Abstract
The microbiota-gut-brain-axis (MGBA) is a bidirectional communication network between gut microbes and their host. Many environmental and host-related factors affect the gut microbiota. Dysbiosis is defined as compositional and functional alterations of the gut microbiota that contribute to the pathogenesis, progression and treatment responses to disease. Dysbiosis occurs when perturbations of microbiota composition and function exceed the ability of microbiota and its host to restore a symbiotic state1. Dysbiosis leads to dysfunctional signaling of the MGBA, which regulates the development and the function of the host’s immune, metabolic, and nervous systems. Dysbiosis-induced dysfunction of the MGBA is seen with aging and stroke, and is linked to the development of common stroke risk factors such as obesity, diabetes, and atherosclerosis. Changes in the gut microbiota are also seen in response to stroke, and may impair recovery after injury. This review will begin with an overview of the tools used to study the MGBA with a discussion on limitations and potential experimental confounders. Relevant MGBA components are introduced and summarized for a better understanding of age-related changes in MGBA signaling and its dysfunction after stroke. We will then focus on the relationship between the MGBA and aging, highlighting that all components of the MGBA undergo age-related alterations that can be influenced by or even driven by the gut microbiota. In the final section, the current clinical and pre-clinical evidence for the role of MGBA signaling in the development of stroke risk factors such as obesity, diabetes, hypertension, and frailty are summarized, as well as microbiota changes with stroke in experimental and clinical populations. We conclude by describing the current understanding of microbiota-based therapies for stroke including the use of pre-/pro-biotics and supplementations with bacterial metabolites. Ongoing progress in this new frontier of biomedical sciences will lead to an improved understanding of the MGBA’s impact on human health and disease.
Keywords: microbiota-gut-brain axis, dysbiosis, aging, stroke, Cerebrovascular Disease/Stroke, Cognitive Impairment, Treatment
Introduction
Microbial organisms colonize the surface of the human body and its mucus membranes. The Human Microbiome Project showed that, even among healthy subjects, the diversity and abundance of these microorganisms evolve throughout the lifespan2 and vary widely depending on the body site. The richest site of microbes is the gastrointestinal (GI) tract3. The collection of all GI microbes and their genetic material are known as the gut microbiota and the gut microbiome, respectively.
Gut microbes consist of approximately 1014 cells that include prokaryotes (bacteria and archaea), eukaryotes (fungi, intestinal protozoa, and parasitic helminths), viruses, and bacteriophages4. The gut bacterial community alone has over 1000 species, 100-fold more genes than the human host4, and an equivalent metabolic capacity to the human liver5. In this review, the term “microbiota” refers specifically to the gut microbiota, primarily the gut bacteria. Intra-individual inter-segment variabilities exist in the microbiota composition with greatest dissimilarity between the cecum and the rectum6. Bacterial load and diversity decrease from the oropharynx to jejunum (upper GI), with Proteobacteria and Firmicutes showing the highest abundance. Bacterial load and diversity increase from the ileum to colon (lower GI), with Firmicutes, Bacteroidetes, and Proteobacteria as the prominent species7. The inter-individual variability of the bacteria is greater than intra-individual inter-segment variability6, regardless of sample collection and processing methods8. Both intra- and inter-individual microbiota variations drive differences in metabolism of dietary molecules and medications, and contribute to disease risk factors9,10.
The human gut microbiota composition is established at birth. Although there may be a prenatal contribution from the placental microbiome, this remains controversial11. During early life, factors including vaginal delivery versus caesarian section (C-section) delivery, breastfeeding versus formula feeding, and exposure to antibiotics are major determinants of gut microbiota composition12. The formation of gut microbiota is essential for development of the metabolic, immune, and nervous systems of the host13. These results have shed light on the importance of the maternal microbiome and its potential effects on the offspring’s long-term risk factors for disease14.
Alteration in the MGBA have been linked to neurodevelopmental disorders (e.g., autism spectrum disorders), neurodegeneration (e.g., Alzheimer (AD) and Parkinson diseases (PD), psychiatric diseases (e.g., depression and anxiety), and stroke15. MGBA pathways can be divided into 1) cellular immune function, 2) direct neural connections, and 3) systemic circulatory factors16. This review will focus on how dysbiosis-induced dysfunction of the MGBA influences stroke risk and outcome. An overview of MGBA structures (Figure 1 and Table 1) and the tools used to study the MGBA are described. As advancing age is a major clinical risk factor for stroke and is independently associated with dysbiosis17, the role of microbiota in age-related changes in the MGBA is highlighted. The contribution of age-related dysbiosis to stroke outcome has not been widely studied in experimental stroke research, as most studies exclusively examined young male mice. The clinical and pre-clinical evidence for a role of the MGBA in stroke pathophysiology is summarized, which highlights the need for consideration of aging and dysbiosis as separate but interdependent biological variables in MGBA signaling in stroke research.
Figure 1.
Illustration of the major participants in MGBA signaling with their anatomical position and serves as a reference throughout this review. MG (microglia), EEC (enteroendocrine cell), Neut (neutrophil), Mac (macrophage), DC (dendritic cell), LP (lamina propria), PP (Peyer’s patch). Three barriers exist between the luminal content and the blood: a mucus layer maintained by goblet cells, a selectively-permeable single layer of enterocytes interconnected with tight junctions, and GBB (composed of enteric glia, pericytes, and endothelial cells). PPs are located along the antimesenteric border of the small intestine and have lymphoid follicles surrounded by APCs and lymphocytes (predominantly B cells). Crypt stem cells at the base of the villi and replenish the epithelium. Goblet cells secrete mucins to maintain the mucus barrier. Paneth cells secrete AMPs that modulate the gut microbiota composition. EECs are differentiated neuroendocrine cells, involved in GI physiology, coordination with the ANS, and feeding behavior. M cells are specialized antigen processing cells in the intestinal PPs capable of transcytosis of luminal antigens. BBB is a highly selective semipermeable barrier comprised of endothelial cells attached by tight junctions, astrocytic end-feet, and pericytes embedded in the vascular basement membrane. CNS lymphatic vessels reside in the dura of meninges with limited access to brain parenchyma through lymphatic protrusions that allows brain-derived antigens to move from the brain to the CSF-filled spaces and cervical lymph nodes. The skull bone marrow houses significant populations of monocytes and neutrophils, with transcriptional signatures distinct from their blood-derived counterparts, that can infiltrate into the brain in response to injury. [Fig 1 artistic rendering by Nicolle R. Fuller, SayoStudio].
Table 1 -.
Summary of the cell types involved in the MGBA
Cells | Description | Link to the Gut Microbiota | Changes with Aging | PMIDs |
---|---|---|---|---|
Crypt Stem Cells | Reside at the base of the villi and continuously replenish the epithelium during physiological shedding or after injury (Figure 1, Gut View) | Microbiota-derived muramyl dipeptide bind Nod2 receptor in Lgr5+ crypt stem cells to increase survival against oxidative stress | • Increase in number but reduced regenerative capacity with aging • Increased differentiation toward PCs and GCs with aging • IL-22 induces their regenerative capacity but IL-22 levels are decreased with aging • SCFAs (butyrate) suppress their proliferation but SCFAs are reduced with aging |
31610128, 28676292, 29141947, 31370376 |
Goblet Cells (GCs) | Secrete mucins (Figure 2a), AMPs, chemokines, and cytokines to maintain the mucus barrier and communicate with local immune cells (Figure 1, Gut View) | • Secrete mucins to maintain mucus layer and stabilize IgA-bacteria complex before phagocytosis by macrophages • Mice deficient in mucus production (e.g., Muc2-/- mouse) have a significantly altered microbiota composition and are more susceptible to colitis, resulting from epithelial erosion, higher expression of immunoglobulins, CD3+ lymphocytes infiltration, and increased levels of proinflammatory cytokines (TNF-a and IL-1b) • Long term supplementation with A. muciniphila, significantly increases the thickness of the colonic mucus layer in mice • GF mice have fewer GCs, a thinner mucus layer, and a higher intestinal permeability than SPF mice |
Reduced number and loss of mucus thickness with aging | 30723224, 33017020, 16831596, 21910166, 30899315 |
Paneth Cells (PCs) | Secretory cells intercalated between the crypt stem cells; PCs do not migrate up the crypt-villus axis and are long-lived (>50 days), unlike other differentiated epithelial cells |
• Th2 cytokines IL4, IL-9, IL-13, and GLP2 released by EECs stimulate PCs to release AMPs. • LP immune cells regulate PCs to secrete AMPs that modulate the gut microbiota composition • Th2 cytokines IL-4, IL-9, IL-13, and EEC-derived GLP2 stimulate the production of AMPs and defense proteins (e.g., lysozyme angiogenin 4, secretory phospholipase A2, and alpha-defensins) by Paneth cells (Figure 2c) • Colonization GF mice with microbiota from SPF mice increases PCs numbers and induces their RegIIIγ expression. |
Increase in number and reduced expression of AMPs (antimicrobial Paneth cell factor (Ang4) and lysozyme) in Paneth cells with aging | 28854151, 34153524, 29883265, 15937933, 28154012, 34153524 |
Enteroendocrine Cells (EECs) | Differentiated neuroendocrine cells, involved in GI physiology and feeding behavior | • Release serotonin to stimulate ENS to initiate gut motility • Express TLRs and sense bacterial components (e.g., LPS) • Synapse with vagal nerve endings (neuropods) |
Decreased gastrin, somatostatin, vasoactive intestinal polypeptide (VIP), substance P, and neuropeptide Y with aging | 33240233, 9852376 |
Microfold (M) Cells | • Specialized antigen processing cells in the intestinal PPs • Distinct from enterocytes with short microvilli and no glycocalyx at their apical membrane (facing the lumen) • Their basolateral membrane (facing the LP) is invaginated to form a pocket that allows direct communication with immune cells • Possess the vesicular machinery to selectively uptake macromolecules from the lumen and secrete those from their basal membrane, a process called transcytosis (Figure 1, Gut View). |
• Antigen transcytosis and mediate IgA production • Specific bacteria (e.g., Bacteroides acidifaciens and Prevotella buccalis) are thought to be indispensable for M cell-dependent maternal IgA synthesis and its secretion into the milk before lactation, suggesting a role of the maternal microbiota in lactation, infant immunity, and other potential long-term effects on offspring health |
• Reduced number of PPs and density of M cells within PPs with aging (and potentially after stroke?) • Antigen processing function of M cells is impaired with aging • Age-related M cell dysfunction is associated with reduced density of a subset of B cells (CD11c+ CCR6+ B cells) responsible for inducing M cells in the PP’s follicle-associated epithelium (FAE) • Passive microbiota transfer from young donors or treatment with bacterial flagellin increases the number of M cells, antigen uptake in the FAE, fecal IgA levels, and co-localization of CD11c+ cells with M cells in aged mice |
34496253, 23360902, 31312204, 34496253 |
Innate Lymphoid Cells (ILCs) | • Belong to the lymphoid lineage and secrete similar inflammatory mediators as T lymphocytes (“lymphoid-like”) yet do not express antigen-specific • receptors and respond to infections quickly (“innate-like”) • ILCs promote migration of DCs to local LNs and regulate T cells through presentation of peptide fragments on their MHC class II |
Microbiota-derived tryptophan metabolites (i.e., indole-3-aldehyde) drives the production of IL-22 by type 3 ILCs, which promotes the expression of AMPs to ameliorate pre-clinical models of intestinal inflammation | Microbiota synthesize less indole derivatives with aging (potential linked to IL-22-dependent function of crypt stem cells?) | 26649819, 23973224, 25902484, 18264109 |
Neutrophils | • Most abundant leukocytes in the blood • Cytoplasmic granules contain enzymes (e.g., lysozyme and collagenase), suicidal proteins (e.g., defensins and cathelicidins), and iron-chelating molecules (e.g., lactoferrin) • Sort-lived (usually hours) and quickly replenished under steady state conditions |
Microbiota regulates neutrophil development and function through TLR-4 and MyD88-dependent pathway to induce the expression of IL-17A and G-CSF | Reduced NETs formation, ROS production, chemotaxis, phagocytosis, degranulation with aging | 31927534, 24747744, 20620114, 27136946 |
Mast Cells | • Tissue-resident cells of the myeloid lineage and mediators of acute allergic response • Cytoplasmic granules of mast cells contain potent inflammatory mediators such as histamine and IgE, which initiate the acute inflammatory response after an injury |
Enterococcus faecalis (given orally) to mice with atopic dermatitis reduces serum IgE levels | Not well defined | 34335160, 32429999, 26959058 |
DCs / Monocytes / Macrophages (MNPs) | • DCs have stellate morphology and are potent APCs that induce differentiation of naïve T cells via their MHC-II • Macrophages are large vacuolar APCs with a high capacity for phagocytosis; Tissue macrophages arise from embryonic progenitors such as yolk-sac or fetal monocytes early during development and give rise to tissue-resident macrophages (e.g., MG or Kupffer cells) that self-maintain throughout life. • Monocytes have unique functions and serve as definitive precursors of DCs and macrophages with overlapping functions and temporal engagement during the immune response • These three cell types are collectively referred to as the mononuclear phagocytes (MNPs) |
• Microbiota induces production of IL-10 by intestinal DCs • Bacterial metabolites (e.g., SCFAs) reduce expression of inflammatory chemokines (e.g., CCL5 and CXCL11) and cytokines (e.g., IL-6 and IL-12p40) after LPS-induced activation of human DCs |
• Reduced TNFa, IL-6, IL-12, IFNs with aging • Impaired chemotaxis, phagocytosis, and production of ROS with aging |
32861719, 24854589, 25033907 |
B Cells | • Bone marrow-derived and distinguished by ability to generate antigen-specific immunoglobulins via an error-prone process called combinatorial genetic rearrangement, antigen driven somatic hypermutation, and affinity maturation • As potent APCs, B cells internalize their target antigen via their B cell receptor (BCR) and present its fragments in an MHC-restricted manner to T or other B cells |
• Microbial antigens affect B cell activation and differentiation via BCRs and TLRs • Moreover, microbiota metabolites can drive the activation of transcription factors in B cells (e.g., 5-HIAA-mediated activation of AHR in regulatory IL-10+ B cells) |
• Reduced number, reduced diversity, reduced IgD-CD27+ subset, increased IgD-CD27- subset, increased TNFa, reduced avidity, increased autoantibody with aging • Exaggerated proliferative response after stimulation with TLR7 and TLR9 ligands • Increased antigen-experienced meningeal B cells with aging |
32213346, 21543762, 18725575, 29522725, |
T Cells | Originate from the bone marrow progenitors, migrate to the thymus for maturation, and redistribute to the periphery | • Segmented filamentous bacteria promote the differentiation of naïve T cells to effector Th17 cells that produce IL-17A, IL-22, and IL-10 • Commensal Clostridium and Bacteroides spp. promote maturation of FoxP3+ regulatory T cells that can suppress Th17 cells and their conversion into IFN-g-producing Th17 cells with direct implications for immunity against intracellular pathogens, systemic inflammation, and neuroinflammation • Gamma delta T lymphocytes (gd T cells), one type of intestinal intraepithelial lymphocytes (IELs), also contribute to local and systemic immunity • The number of IL-17A+ gd T cells in the intestinal LP and the liver of GF mice is lower than in SPF mice, a process mediated by CD103+ DCs and IL-10+ Treg cells |
• Reduced naïve T cells, reduced Treg, reduced CD40L, increased inflammatory molecules, increased Th17:Treg ratio, decreased IL-2 with aging • Increased memory, senescent, CD28−, PD-1+, CTLA-4+ T cells with aging |
23378581, 34341528, 11588048, 29466753, 34341528 |
MG / CNS BAMs | • Innate CNS-resident macrophages, endowed with memory-like functions for context-dependent responses • They are derived from the yolk sac and begin to colonize the CNS at around the 4th gestational week in humans (embryonic day 7.5 in mice), long before other glia cells arise • MG are long-lived and maintain their tissue-specificity via their potential to self-renew • MG participate directly in the neuroimmunology of CNS diseases, including AD, PD, schizophrenia, multiple sclerosis, and stroke |
• MG from GF mice display a hyper-ramified morphology and a dampened response to inflammatory stimuli • Abnormal MG phenotype in GF mice is reversible via microbiota reconstitution from SPF mice or supplementation with microbial metabolites (e.g., SCFAs) • Tryptophan-derived indole-based ligands activate the MG AHR to suppress the activation of the NF-kB pathwayMicrobiota-dependent activation of AHR reduces VEGFB expression by MG and increases TGF-a secretion by astrocytes, which leads to dampened astrocyte-mediated neuroinflammation through a SOCS2-dependent pathway (Figure 3a) |
• Increased activation state (e.g., increased CD45 and MHCII while decreased P2RY12) and reduced phagocytic capacity with aging • Peripherally-sourced DCs/Macrophages increase in the brain with aging Increased CD38+MHCII+ BAMs with aging |
33261619, 29752550, 31585077, 29881376, 33556248, 26030851, 29769726, 27158906 |
TOOLS FOR STUDYING THE GUT MICROBIOTA
Metabolome Analysis:
Low-molecular-weight molecules (metabolites) such as vitamins, fatty acids, amino acids, and bile acids are regulated by the microbiota and the host and serve as important MGBA signaling molecules in aging and stroke (see corresponding sections). Due to their ability to detect a wide range of metabolites, mass spectrometry (MS)-based platforms such as gas chromatography-MS (GC-MS), liquid chromatography-MS (LC-MS), and capillary electrophoresis-MS (CE-MS) are increasingly utilized in metabolomics analysis of MGBA signaling.
Microbiome And Metagenome Analysis:
The conserved 16S ribosomal RNA (16S rRNA or 16S) gene is used to identify and compare the bacterial composition. The 16S gene has conserved regions that identify bacteria versus non-bacteria (with the exception of some 16S primers that also detect Archaea) and has variable regions (V1-V9) to distinguish individual bacteria taxa. After taxonomic assignment, the bacterial diversity of samples is compared using alpha (within-sample) and beta (between-sample) diversity metrics. Alpha diversity is a qualitative measure of the composition of a microbial community within a site or within a sample that increases with increased richness (the number of taxonomic groups and increased evenness (the relative abundance of the existing taxonomic groups) 18,19. Beta diversity describes the species diversity between two or more microbial communities from different samples using variety of phylogenetic metrics (e.g., weighted and unweighted UniFrac20) and non-phylogenetic (e.g., Bray-Curtis or Jaccard) metrics21. 16S data does not provide direct insights into the biochemical functions of the gut microbiota. PICRUSt (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States) is an example of a computational approach to predict the functional composition of a metagenome22. Integrated metagenomic and metabolomic analyses of microbial samples (a powerful example of multi-omics) can be used to identify the link between key bacteria and translationally important metabolic and immune pathways in aging and stroke (discussed later).
Experimental Models For Studying The MGBA:
Antibiotics:
Although a useful tool to manipulate the host gut microbiota, antibiotic administration can have off-target effects on microbiota at other sites (e.g., lungs, skin, eyes, oropharynx, etc.). Some antibiotics can also elicit microbiota-independent changes in host metabolites and phagocytic clearance of pathogens by reducing cellular respiration in immune cells23. Antibiotics can alter cellular metabolism and lead to hyperglycemia, increased adiposity, and insulin resistance, known risk factors for stroke24. Colonocytes adapted their preference for SCFAs as an energy source toward a preference for glucose via increased glucagon-like peptide 1 (GLP-1) signaling in response to a sudden reduction of luminal SCFAs following antibiotic treatment25. The effects of antibiotics depend on the antimicrobial coverage, dose, duration, and host factors, such as age, sex, diet, strain, and presence of other pathologies26, which must be carefully validated for the specific experimental conditions.
Fecal Microbiota Transplant (FMT):
Successful FMT requires a diligent design and validation of several parameters including age, sex, strain, vendor, disease, and treatment status of both donors and recipients. Collection methods, time-to-freeze versus fresh use, freeze-thaw cycles, oxygen exposure, and preservatives must be evaluated and reported. An important clinical and pre-clinical consideration in FMT use is that all microbial content (bacterial and non-bacterial) is transferred. Slowly degradable metabolites, viral, fungal, and parasitic genes are also introduced into the recipient. These non-bacterial entities pose a clinical risk of transferring infections and can be a source of experimental confounders.
In Vitro Systems such the gut-on-a-chip27 or SHIME (Simulator of the Human Intestinal Microbial Ecosystem28) are used to investigate the direct implication of the microbiota on gut homeostasis. Purified intestinal bacterial cultures derived from a single healthy donor’s stool have been used to treat recurrent hypervirulent C. difficile infection in patients who had failed antibiotic treatments29. In the same study, recipients remained symptom-free at six months after FMT and their stool samples demonstrated over 25% similarity to the synthetic stool substitute29. In vitro approaches may effectively address the FMT’s clinical safety concerns30. Human intestinal organoids grown from induced pluripotent or embryonic stem cells are used to study clinical pathologies such as response to C. difficile toxin31. These organoids are sterile (unless bacteria are introduced), have a functional epithelial barrier, and are composed of lysozyme- and mucin-producing cells that resemble Paneth and goblet cells (see Table 1)32. Organoid models provide experimental control over the oxygen and nutrient levels. To accelerate drug development, engineered microchips containing living human cells that reconstitute organ-level functions have been developed33,34.
Gnotobiotic Models:
Germ free (GF, devoid of detectible bacteria) or gnotophoric (one or a few known bacteria) models have revealed microbiota’s role in the development of the immune and nervous systems. GF models have limitations, such as defective immune responses35, reduced blood-brain barrier integrity, and altered microglial maturation and function, and therefore may be less translationally relevant for human studies. Specific changes observed in GF mice are summarized in Table 2. The artificial nature of animals raised in vivaria may also lead to conclusions with less translational relevance. Other model organisms, including studies in Drosophila and Zebrafish have explored the MGBA and have been recently reviewed36. There are also differences between breeders and mouse strains that impact MGBA signaling37. Importantly, baseline differences in MGBA immunity, neurochemistry, and behavior in GF mice are potential experimental confounders in preclinical studies of the role of MGBA in stroke.
Table 2 -.
Summary of MGBA changes seen in “Dirty” Wild-Caught Mice, Wild Type Mice (SPF), and Germ Free (GF) Mice.
“Dirty” Wild-Caught Mice | Wild Type Mice (SPF) | Germ Free (GF) Mice |
---|---|---|
Most CD8 T cells have effector memory phenotype (Beura et al. 2016) | Most CD8 T cells have naïve T cell phenotype; SPF mice have >20% mortality when exposed to pet store mice (Beura et al. 2016) | • Reduced plasma SCFAs (Erny et al. 2015) (Perry et al. 2016) • Reduced plasma indole-based tryptophan metabolites (indoxyl-3-sulfate)(Wikoff et al. 2009) • Smaller PPs with a reduced CD4+ Th17 and IgA-secreting plasma cells (reversible by SCFAs supplementation) (Mazmanian et al. 2005) (Hapfelmeier et al. 2010) • Colonization of GF mice with SPF microbiota restores IgA • Devoid of LP T cells (reversible by FMT from SPF mice) (Umesaki et al. 1993) • Higher network connectivity, less structural organization in the brain (Aswendt et al. 2021) • Lower intestinal 5HT and GLP-1; Eat less than SPF mice (Figure 2c)(Modasia et al. 2020) • Lower 5HT in GF increased by spore-forming bacteria colonization • Increased intestinal sympathetic activation (decreased by SCFA-producers) • Reduced intestinal transit rate (increased by FMT from SPF mice via 5HT4R) • Increased BBB permeability and reduced expression of tight junctions (reversible by FMT from SPF mice) (Braniste et al. 2014) • MG display a hyper-ramified morphology and a dampened response to inflammatory stimuli (reversible by FMT from SPF mice or SCFAs supplementation) (Erny et al. 2015) • Behavioral abnormalities: Exaggerated HPA stress response (reversible by FMT from SPF mice); motor hyperactivity and reduced anxiety (Sudo et al. 2004) (Diaz Heijtz et al. 2011) • Proper controls: WT littermates, co-housing controls, cross-facilities, and matched strain backgrounds (Stappenbeck and Virgin 2016) |
Enormous advances in the tools available to study the role of MGBA in stroke have been made. However, potential pitfalls make interpretation of existing evidence and design of future studies challenging. These pitfalls include differences in the anatomy, immunology, microbiota composition, multi-comorbidities including aging, sample preparation, bioinformatics approaches, and thresholds for distinguishing “correlation versus causation” in MGBA research38. For instance, the role of gut virome or bacteriophages of the microbiome in stroke have not been investigated. Attempts for selective bacterial colonization remain artificial, neglecting the effects of complex microbial signaling such as quorum sensing and biofilm39. Despite inherent limitations, gnotobiotic models remain valuable in establishing causality in MGBA studies of stroke, discussed later in this review.
THE MICROBIOTA-GUT-BRAIN AXIS
Understanding the integrated anatomy and physiology of cells and specialized structures of the MGBA is critical to our understanding of its role in health and disease. The purpose of this section is to provide a brief overview of MGBA components (Figure 1), description of key cells and their link to the gut microbiota (Table 1).
Intestinal Barrier And Permeability:
Three barriers exist between the intestinal lumen and the blood: a mucus layer maintained by goblet cells, a selectively-permeable single layer of enterocytes interconnected with tight junctions, and a gut-blood barrier (GBB) composed of enteric glia, pericytes, and endothelial cells40 (Figure 1). The intestinal villi and crypts, finger-like projections and infoldings, are composed of enterocytes that absorb nutrients. Enterocytes, goblet, Paneth, enteroendocrine, crypt stem, and microfold (M) cells41 maintain a homeostatic barrier with the luminal microbiota (Table 1 and Figure 2a). Secretion of mucins, antimicrobial peptides (AMPs), secretory IgA into the lumen, and expression of tight junction proteins (e.g., E-cadherin, Figure 2b)42 allow for selective permeability to nutrients while preventing translocation of bacterial and deleterious molecules.
Figure 2.
Representative fluorescence in situ hybridization (FISH) showing close proximity of bacteria to gut epithelium in naïve age mice (red arrow), compared to young mice (green arrow) (a, top two panels, Red: bacteria (non-specific FISH probe), Blue: DAPI, 10x magnification). Representative immunohistochemistry showing a loss of mucin secretion (pink arrows) in naïve aged mice (2a, bottom two panels, brown: MUC-2 stain, blue: hematoxylin, 40X Magnification). Representative immunohistochemistry showing the expression of intestinal E-cadherin tight junction protein (orange arrow) by intestinal epithelial cells (b, Cecum, brown = E-cadherin, blue = hematoxylin, 10/40x magnification). Signaling network between afferent vagal fibers, EECs (neuropods), Paneth cells, and LP Th2 lymphocytes. Colonization of GF mice with microbiota from SPF mice or even a single bacteria (e.g., Bacteroides thetaiotaomicron) can restore the serotonin and GLP1 production in EECs in GF mice. Th2 cytokines IL-4, IL-9, IL-13, and EEC-derived GLP2 stimulate the production of AMPs by Paneth cells. EECs express TLR-2 and TLR-6 that sense bacterial lipopolysaccharide (LPS) to activate the canonical NF-kB pathway. Afferent sensory signals propagate toward the brainstem and efferent (CNS-originated) signals travel to the intestinal neuropods in the order of milliseconds to regulate the release of hormones, including GLP1, CCK, ghrelin, and serotonin (c). [created with BioRender.com]
Gut-Associated Lymphoid Tissues (GALTs) include the lamina propria (LP), mesenteric lymph nodes, and Peyer’s patches (PPs) (Figure 1). GALTs contain immune cells that coordinate the host’s local and systemic defense to intestinal insults. The LP is a thin layer of loose, non-cellular connective tissue beneath the epithelial layer with a high amount of immune cells and nerve endings. PPs are dome-like structures along the antimesenteric border of the small intestine with lymphoid follicles surrounded by antigen presenting cells (APCs) and lymphocytes (predominantly IgA-producing plasma cells). The follicle-associated epithelium of PP has a thin mucus layer and M cells for the transport of luminal antigens to the LP. The role of GALTs in aging and stroke is discussed in the following sections.
Enteroendocrine Cells (EECs) are differentiated neuroendocrine cells involved in GI physiology and feeding behavior. Enterochromaffin cells, the largest subset of EECs, synthesize over 90% of the entire body’s serotonin (5-hydroxy tryptamine, 5HT). Some EECs (neuropod cells43) directly synapse with enteric nerve endings and secrete 5HT to initiate a reflex that regulates water balance in the lumen via the submucosal plexus and to trigger gut peristalsis via the myenteric plexus (Figure 1)44. Neuropods sense mechanical, thermal, and chemical signals from the lumen and convert these inputs into electrical pulses. Neuropod-generated electrical pulses can be transmitted across the serotonergic synapses formed by the neuropods and afferent vagal fibers. Afferent sensory signals propagate toward the brainstem and efferent (CNS-originated) signals travel to the intestinal neuropods in milliseconds to regulate the release of EEC hormones and neurotransmitters, including 5HT, somatostatin, neuropeptide Y, cholecystokinin (CCK), and GLP-1 (Figure 2c)45.
5HT is also a CNS neurotransmitter involved in the regulation of mood, memory, and cognition, even though it is unclear whether sufficient concentrations of gut-produced 5HT reach the CNS46. 5HT does not cross the BBB47. The effects of 5HT, whether gut-derived, brain-derived, or transported along the MGBA by other cells is a major pathway that influences brain immunity. Gut-derived 5HT produced by EECs is stored and released from platelets (reviewed in48) and other immune cells. DCs express multiple 5HT-activated receptors, respond to 5HT by secreting IL-6, store 5HT and upon ATP-mediated calcium influx, can release 5HT and other cytokines to activate B and T lymphocytes49. 5HT drives anti-inflammatory macrophage polarization via 5HT2B and 5HT750. Microbiota-regulated 5HT can have direct and indirect effect on immune response after brain injury during which platelets, CNS border-associated macrophages (BAMs), and other infiltrating myeloid cells capable of storing and releasing 5HT are involved.
Gut microbiota regulate hunger and satiety by controlling EECs. EECs express chemosensory and pattern recognition receptors (e.g., toll-like receptor (TLR)-2 and TLR-6) that detect bacterial lipopolysaccharide (LPS) to activate the canonical NF-kB pathway (Figure 2c)51, indicating that EECs can directly participate in local immune responses in addition to their active role in 5HT-mediated immune regulation and feeding behavior. The transcriptomic signature of EECs among obese subjects can discriminate between individuals with or without diabetes. Specifically, obese patients with diabetes showed reduced GLP-1 cell differentiation and proglucagon maturation as well as reduced plasma GLP-152. EECs and their regulation of 5HT and GLP-1 signaling are a prime example of a specialized intestinal cell that can serve as a sensor for microbiota- and CNS-derived neurochemical signals. This allows for the coordination of GI secretory function, systemic immunity, feeding behavior, obesity and diabetes, known risk factors for stroke. The role of serotonergic system in stroke outcomes is an area of active clinical investigation53. Changes in EECs and other specialized epithelial cells with aging, a major stroke risk factor, are summarized in Table 1.
The Lymphatic System is crucial for establishing systemic immune responses by serving as the “exit conduits” for activated APCs to reach the lymph nodes (LNs) and non-tissue-resident immune cells54. The presence of meningeal lymphatics was described by an early Italian anatomist (Paolo Mascagni) over 200 years ago55. Kipnis56 and others used direct microscopy to confirm the presence of an intracranial meningeal lymphatic network in mice, later reported in non-human primates and humans57. Although these lymphatic vessels reside in the dural layer of meninges without direct access to the parenchyma, limited access through lymphatic protrusions allows brain-derived antigens to move from the sub-pial brain into the CSF-filled spaces to cross the arachnoid mater and reach the dural lymphatics58 (Figure 1). Mouse and human meninges house IgA-secreting plasma cells that originate in the intestines. These cells increase with aging and after breakdown of intestinal barrier59, which is seen after stroke. These findings indicate that brain-derived antigens can interact with meningeal APCs and IgA-secreting plasma cells to mount an antigen-specific adaptive immune response that requires the presence of the gut microbiota.
The Blood-Brain Barrier (BBB) is a semipermeable barrier comprised of endothelial cells, tight junctions, astrocytic end-feet, and pericytes (Figure 1). Microbiota and its metabolites interact with the BBB60. During inflammation, BBB permeability, expression of adhesion molecules, and brain uptake of neutral amino acids are increased61. BBB breakdown allows circulating microbiota-derived metabolites (e.g., tryptophan metabolites, bile acids, and SCFAs) to gain direct access to CNS myeloid cells and astrocytes, essential regulators of BBB62. Exploiting the regulatory role of the microbiota metabolites is an active area of MGBA research.
Blood-CSF Barriers are important sites for peripheral-CNS communication63. The choroid plexus and other structures maintain the blood-CSF barriers (Figure 1). Blood flow to choroid plexus can be regulated by 5HT64, which can be a microbiota-dependent pathway as discussed above. Stroke reduces blood flow to the choroid plexus, which leads to increased permeability of the blood-CSF barrier65. Blood-CSF barriers contain transmembrane proteins (e.g., ATP-binding cassette transporter and solute carriers), crucial for exchanging microbiota-derived metabolites such as indoxyl sulfate (a tryptophan metabolite) and trimethylamine-N-oxide between these fluid compartments66. The role of these metabolites in stroke are discussed below.
Cerebral Blood Flow (CBF) plays an important role in BBB and blood-CSF physiology. Increased CBF was seen in mice fed a chronic ketogenic diet, which increased A. muciniphila and Lactobacillus and reduced Desulfovibrio and Turicibacter, demonstrating that specific shifts in microbiota can regulate CBF67. The role of nitric oxide synthase (NOS) and arginase in NO-dependent regulation of CBF in stroke is well established68. Arginase and NOS compete to degrade L-arginine, and plasma levels of L-arginine are directly regulated by the microbiota69. Microbiota-dependent arginine and tryptophan metabolism that influence NO-mediated70 or 5HT-mediated regulation of CBF64, respectively, are clinically relevant pathways in stroke or cerebral vasospasm pathologies.
CELLULAR IMMUNE PATHWAYS:
Immune Surveillance In The LP:
Immune tolerance is defined as the absence of an effector response to an immuno-potent antigen71. In addition to M cells (Table 1), tolerogenic (CD103+) DCs directly sample luminal antigens (Figure 1) and descend from the epithelium to present their antigen to naïve T cells, inducing their differentiation into anti-inflammatory Treg cells. Treg cells travel to the LP to secrete anti-inflammatory cytokines (e.g., IL-10 and TGF-β) to maintain immune quiescence71. Gut microbiota regulate the mucus layer and antimicrobial function of goblet and Paneth cells (Table 1), and plays an active role in inducing LP immune cells, particularly LP lymphocytes (Table 2). The role of microbiota in LP immunity and inflammation has been extensively reviewed elsewhere72.
CNS Myeloid Cells:
The gut microbiota influences microglia (MG) biology throughout life (Tables 1 and 2). Within minutes after injury, MG sense DAMPs (e.g., ADP and ATP via their P2Y12 receptor) to initiate chemotaxis to the site of injury. Upon activation, MG retract their processes to transform into an amoeboid morphology and begin coordinating with other CNS cells and peripherally-sourced immune cells. Microbial metabolites directly regulate MG-mediated neuroinflammation62,73. Tryptophan-derived indole-based ligands activate the MG aryl hydrocarbon receptor (AHR, a regulator of immune differentiation and neuroinflammation) to suppress the activation of the NF-κB pathway. Microbiota-dependent activation of AHR reduces VEGF-β expression by MG and increases TGF-α secretion by astrocytes to dampen astrocyte-mediated neuroinflammation through a SOCS2-dependent pathway (Figure 3a)74. CNS BAMs, which line the meninges, choroid plexus, and perivascular spaces (Figure 1), are CNS-resident myeloid cells, distinguished from MG by higher expression of CD38 and MHC-II, while lacking typical MG markers (Tmem119 and P2RY12)75,76. The choroid plexus is a site high cerebral blood flow and a site of recruitment for inflammation-resolving immune cells from the peripheral circulation77–79. At blood-CSF interfaces such as the choroid plexus and meninges, BAMs and other APCs continuously monitor and filter brain-derived antigens at steady state and after stroke.
Figure 3.
Microbiota-dependent activation of the MG AHR reduces VEGFB by MG which increases TGF-a secretion by astrocytes, which in turn dampens astrocyte-mediated neuroinflammation (a). Age-related decrease in protective bacteria (e.g., Akkermansia or SCFA-producers) can lead to reduced intestinal barrier function, which activates DC to stimulate gd T cells, increasing their migration to the brain and enhancing the production of IL-17, leading to the additional recruitment of neutrophils into the aged brain after stroke (b). [created with BioRender.com]
NEURAL PATHWAYS:
Direct neural pathways link the GI tract and the CNS through the autonomic nervous system (ANS). ANS divisions are the sympathetic, parasympathetic, and enteric* nervous systems. Sympathetic and parasympathetic divisions contain afferent (ascending) and efferent (descending) fibers consisting of preganglionic and post-ganglionic neurons. Acetylcholine (ACh) is the main neurotransmitter of all presynaptic neurons as well as post-synaptic parasympathetic neurons. Norepinephrine (NE) is the effector transmitter of post-synaptic sympathetic neurons. In addition to ACh, important signaling molecules such as neuropeptide Y (NPY), vasoactive intestinal peptide (VIP), and ATP can act as neurotransmitters of the ANS as well as chemoattractants for immune cells (e.g., monocytes80).
The Sympathetic System innervates nearly all body tissues. Its activation increases blood pressure, heart rate, and glycogenolysis, and dampens pathways involved in digestion81. Bacterial metabolites affect sympathetic signaling, with an overall suppressive effect of the microbiota over ascending sympathetic signaling (Table 2).
The Parasympathetic System regulates feeding, anxiety, arousal, memory, mood, and also senses microbial metabolites82. The parasympathetic division exerts its effects primarily via the vagus nerve (VN, cranial nerve X, innervating the upper GI to proximal colon) and pelvic (innervating the distal colon to anus) nerves. The VN regulates intestinal motility by activating the ENS motor neurons in the myenteric plexus. The VN carries over 70% of the parasympathetic fibers and innervates thoracic and abdominal organs, including the entire GI tract. VN innervates the muscularis propria, LP, and epithelium of the GI tract and can directly sense luminal signals83. In cultured intestinal organoid models, bacteria-derived SCFAs are sensed by EECs, resulting in a calcium-dependent 5HT release from the EECs that is relayed to the adjacent vagal nerve endings that express 5HT3R (type 3 serotonin receptor)84. These findings suggest that bacterial metabolites can trigger neurochemical signals that ascend to the vagal nuclei in the brain stem in milliseconds with implications in feeding, arousal, and mood-related behaviors.
Importantly, VN stimulation (VNS), used in management of treatment-resistant depression85, may be neuroprotective after stroke86. Both VN-dependent and T cell-dependent cholinergic signaling regulate Paneth cell’s AMP production and epithelial cell proliferation87. VNS activates the α7 nicotinic ACh receptor, an important mediator of the cholinergic anti-inflammatory pathway involved in the response to stroke86. Activation of the α7 nicotinic ACh receptor reduced pyroptosis, a form of programmed cell death86 and upregulated BBB tight junction proteins (e.g., claudin-5 and occludin)88. Taken together, vagal cholinergic signaling is involved in both top-down (anti-microbial and intestinal motility) and bottom-up (BBB permeability and cell survival) signaling along the MGBA with direct implications in response to stroke.
The Enteric Nervous System (ENS) is an extensive, self-contained web of over 100 million enteric neurons and enteric glial cells (EGCs). The ENS is composed of the myenteric (Auerbach) plexus that regulates peristalsis and the submucosal (Meissner) plexus that regulates water and electrolyte movements89. The ENS neurotransmitters include ACh, nitric oxide (NO, released from enteric neurons and EGCs90), and 5HT91, which can be released by EECs in response to increased microbiota metabolites (e.g., SCFAs)84. Microbiota-derived tryptophan metabolites increase intestinal motility through activation of AHR in enteric neurons92. These findings suggest a microbiota-ENS communication that can convert luminal metabolites into neurochemical signals that regulate intestinal physiology and may play a role in post-stroke complications such as ileus (discussed later).
SYSTEMIC CIRCULATION (HORMONES, CYTOKINES, AND METABOLITES):
The Hypothalamic-Pituitary-Adrenal (HPA) Axis regulates the body’s response to psychological and physical stressors. Stress-induced activation of the HPA axis increased GI permeability through sympathetic activation of beta-adrenergic receptors, inducing intestinal Th17 cells, and altering the gut microbiota composition in mice93,94, suggesting a top-down regulatory role of the HPA axis on the microbiota composition that can contribute to stroke-induced dysbiosis. In addition to HPA’s neurohormonal role, host circadian rhythm also affects MGBA at steady state and in response to stroke95. For example, a study using circadian mutant ClockΔ19/Δ19 gene knockout mice revealed that a functional circadian signaling is required for the beneficial effects of the gut microbiota on recovery after myocardial infarction96, however this has not yet been examined in models of stroke.
Cytokines:
The gut microbiota can drive CNS pathology both through its metabolites and by modulation of circulatory cytokines. Dysbiosis can lead to increased circulatory proinflammatory cytokines such as IL-1β, IL-6, TNF-α, and IFN-γ, disruption of tight junctions and increased permeability across the intestinal epithelial barrier, GBB, and BBB97. Dysbiosis-induced disruption of these barriers provides a direct bidirectional gateway for the brain-derived antigens to trigger a systemic immune response and for microbiota-derived metabolites to access the CNS immune compartment. It is difficult to discern the direct effects of metabolites or cytokines on the CNS from their indirect effects via immunological and peripheral neural pathways98. Bile acids, SCFAs, and tryptophan metabolites represent well-studied classes of microbiota metabolites that mediate the MGBA communications and will be discussed under the MGBA and Stroke section of this review.
AGING AND THE MGBA
Aging is associated with a loss of nutrient absorption, anorexia, malnutrition, and frailty99. Aging increases the risk for cancer, neurodegenerative, cardiovascular, and cerebrovascular diseases. The underlying mechanisms that contribute to biological aging include increased DNA damage, telomere shortening, impaired autophagy, and immunosenescence100,101. Recently, the gut microbiota has emerged as a key mediator of the aging process and in age-related pathologies.
Aging And The Intestinal Barrier:
Intestinal barrier function deteriorates with aging, which correlates with an increased rate of GI infections in the elderly102. The loss of barrier function is due to an age-related decrease in tight junction proteins and mucus production103, which can be regulated by the microbiota. For example, A. muciniphila abundance is lower in patients with age-related disorders than in healthy age-matched controls104. A. muciniphila supplementation ameliorates age-related reductions in colonic mucus thickness105. A probiotic cocktail of Lactobacillus and Enterococcus strains isolated from healthy infants given to aged mice with dysbiosis (induced by a high fat diet) reduced intestinal permeability through increased bile salt hydrolase activity, inducing a taurine-dependent expression of tight junctions (Zo-1 and occludin)106. These findings suggest that age-related loss of intestinal barrier function can be reversed by microbiota-based therapies.
Aging And Specialized Intestinal Epithelial Cells:
IL-22 secretion, which stimulates the regenerative capacity of crypt stem cells, is reduced with aging107. IL-22 production by the intestinal ILCs can be induced by microbial metabolites such as indole-3-carboxaldehyde, a tryptophan-derived AHR ligand108,109. The microbiota’s capacity to synthesize indoles progressively decreases with aging110. These findings suggest that the lower availability of indole-based metabolites seen with aging reduces the ILC3-dependent production of IL-22, which decreases the regenerative capacity of crypt stem cells. SCFAs (e.g., butyrate) can suppress stem cell proliferation via a FoxP3-dependent HDAC inhibition111. Fecal SCFAs decrease with aging112, which may explain the age-related increase in crypt stem cells.. Aged mice have reduced thickness of the mucus barrier, impaired M cell antigen processing, reduced specialized plasma cells113. Cell-specific changes in the specialized epithelial cells with aging are included in Table 1. As a result these age-related changes and stroke-induced dysbiosis, the microbiota can encroach upon the epithelium and initiate inflammation (Figure 2a).
Aging And The Immune System
Aging is associated with remodeling of the immune system and increased low-grade inflammation, termed immunosenescence and inflammaging, respectively. Immunosenescence is characterized by a decline in immune efficacy to fight infections, impaired effector responses to novel antigens, increased autoimmunity, and poor wound healing. Inflammaging, often viewed as a consequence of immunosenescence, is characterized by increased serum levels of proinflammatory cytokines including IL-1β, IL-6, IL-8, and TNF-α and decreased anti-inflammatory cytokines like IL-10101.
Microbiota And Age-Related Changes In Innate Immunity:
Consistent with reported increased proliferation and tissue-redistribution of myeloid cells with aging, we have shown that peripherally-sourced APCs significantly increase in the brain with advancing age114. This myelocytic accumulation in the brain positively correlated with age-related changes in the gut microbiota (decreased Akkermansia) and behavioral deficits114. Although the proportion of neutrophils in circulation does not appear to be affected, their functions (chemotaxis, phagocytosis, production of ROS) are impaired with aging101,115. The microbiota drives neutrophil aging via TLR and MyD88 signaling, and depletion of the microbiota (in GF or antibiotic-treated mice) reduced the number of circulating aged neutrophils (CXCR4highCD62Llow) and improved survival in animal models of endotoxin-induced septic shock116.
Microbiota And Age-Related Changes In Adaptive Immunity:
Age-related changes in T cell populations include decreased numbers of naïve T and Treg cells and increased CD8+ memory and Th17 cells117. Age-related changes in the B cell include a decline in B cell lineage precursors117. With aging, peripheral B cell phenotypes shift away from naïve B cells toward antigen-experienced B cells with restricted clonal diversity with impaired capacity to produce novel high-affinity immunoglobulins118. Age-associated subsets of B cells have been described as a population with high expression of CD11c and the transcription factor T-bet. These B cells accumulate in the blood and meninges with age and can undergo an exaggerated proliferative response after stimulation with TLR7 and TLR9 ligands119,120.
Studies have shown that the marked shift from lymphocyte proliferation to myeloid cell proliferation with aging in SPF mice is abrogated in GF mice121. There is an expansion of B cells in GF mice, suggesting a suppressive role of microbiota on B cell proliferation in the bone marrow. Additionally, A. muciniphila supplementation in an accelerated aging model (Ercc1−/Δ7 mice with impaired DNA repair protein ERCC1) decreased the number of activated CD80+CD273− B cells in PPs105. These findings implicate the gut microbiota in the regulation of lymphocyte proliferation and suggest a possible treatment opportunity for lymphocyte-dominant disorders (e.g., autoimmune diseases) and as a potential adjunctive therapy used in combination with approved B cell depletion medications.
Aging And CNS Immunity:
Age-related changes in CNS immune cells are summarized in Table 1. Microbiota depletion using antibiotics or in GF models have demonstrated a detrimental role for the microbiota in AD122. FMT from WT controls into APPswe/PS1dE9 transgenic mice (a model of AD) improved cognitive deficits, reduced brain deposition of amyloid-β (Aβ), and decreased neuroinflammation (reduced COX-2 and CD11b expression) in the brain. These effects were associated with increased fecal SCFAs (butyrate)123. GF AD mouse models showed reduced Aβ deposition. Supplementation of SCFAs in GF AD mice upregulated MG expression of the ApoE gene and increased Aβ deposition, suggesting that microbiota-derived SCFAs promote Aβ deposition, likely by regulating MG phenotype and phagocytosis124. Lymphatic drainage of CNS antigens is decreased with aging125,126, suggesting that exposure of brain-derived antigens to BAMs and other APCs may be prolonged in aged compared to young stroke. FMT from aged donors into young recipients increases MG activation (decreased P2RY12 and increased Iba-1 and MHCII expression) and systemic neuroinflammation127,128. Moreover, GF mice that received FMT from aged WT mice show decreased fecal SCFAs (acetate, propionate, and butyrate) and demonstrate depressive-like behavior, impaired short-term memory, and impaired spatial memory over the 3 months following FMT129.
Dysbiosis has emerged as a therapeutically-targetable contributor to immunosenescence130. Rejuvenation procedures (co-housing, serum-injections, and heterochronic parabiosis) showed that the microbial community and intestinal immunity of aged mice becomes comparable to those of young mice131. Metagenomic data implicated that a higher abundance of Akkermansia and butyrate-producers were responsible for the immune rejuvenation seen in aged mice131. These findings suggest that microbiota can drive age-associated neuroinflammation, independently of the host’s age, and can contribute to post-stroke inflammation127,132.
Aging And MGBA Neural Pathways
Up to 50% of the aging population experiences chronic constipation and GI-related issues. A seven year clinical study of over 3 million veterans found that higher rate of constipation is independently associated with a higher risk of all-cause mortality and ischemic stroke133. Studies in rodents have demonstrated that aging reduces intestinal smooth muscle contractility134 and its neural innervation135 and slows intestinal transit time136, suggesting age-related dysfunction of the ANS that can be driven by the microbiota and exacerbate post-stroke complications such as ileus.
Microbiota And Age-Related Changes In Sympathetics/Paraympathetics:
Age-related changes in the ANS function include decreased neurotransmitter function, leading to diminished regulatory activity (e.g., CBF regulation) and poorly coordinated autonomic discharge (e.g., bladder function)137. Resting sympathetic tone increases with aging, which is correlated with increased plasma NE and decreased sensitivity of the catecholamine and cholinergic receptors137, which implies an age-related cholinergic dysfunction of CAIP after stroke (discussed above). FMT from spontaneously hypertensive rats into normotensive rats increases sympathetic activity, blood pressure, and neuroinflammation138, suggesting that microbiota can mediate sympathetically-driven pathophysiology seen with aging and in stroke.
Microbiota And Age-Related Changes In ENS:
ENS neurons undergo age-related neurodegeneration in animals139. The loss of enteric neurons with aging reduces the density of nerve fibers in the intestinal mucosa140. Aged enteric neurons are swollen and dystrophic and exhibit lipofuscin accumulation and higher ROS production in the myenteric plexus141. Age-related reductions in EGCs, proportional to loss of enteric neurons, has also been described142. Although, communication pathways are present between the microbiota and ENS, the direct contribution of age-related dysbiosis to ENS neurodegeneration has not been elucidated.
In summary, all components of the MGBA undergo significant age-related alterations, many of which can be directly influenced by or even driven by the microbiota. Given these observations and the fact that stroke is a disease of the aged, studies of stroke pathophysiology and outcomes must consider the age-associated background in the host and the microbiota, including dysbiosis, GI dysfunction, immune remodeling, and other existing CNS or systemic pathology.
STROKE AND THE MGBA
Stroke is a leading cause of death and long-term disability143. Ischemic stroke, secondary to the loss of cerebral blood supply caused by thrombosis or embolism143 is the most common subtype, and will be the focus of this section. Survival after ischemic stroke has improved due to advances in its acute management, intravenous thrombolytics (Alteplase (tissue plasminogen activator, TPA) and Tenecteplase (TNK)) and endovascular thrombectomy144. Due to the exclusion criteria driven by timing, risk of hemorrhage, and demand for specialized resources, fewer than 10% of stroke patients receive these treatments145. The poor long-term functional outcome of stroke survivors is a major driver of loss of quality of life and the increasing socioeconomic burden of stroke146.
Stroke Risk Factors And The MGBA:
The rate of infection after stroke is ~30%, and post-stroke infections are associated with poor outcomes147. Up to 50% of stroke patients also experience GI-related complications, including GI bleeds, loss of peristalsis, and dysbiosis, causing prolonged length of hospital stay, higher mortality, and poorer recovery148. The signaling cascade induced by stroke does not stop at perturbing GI homeostasis and the microbiota. A wave of bottom-up signaling propagates toward the CNS, contributing to the subacute and chronic phases of response to stroke. This section will describe how dysbiosis and aging affect MGBA signaling to play a detrimental, yet potentially reversible, role in stroke outcomes. Although stroke is a disease of aging, the majority of preclinical research has been conducted in young animals.
Multiple studies have shown that infarct volumes are smaller after transient MCAO in aged mice compared to strain-matched young mice, even with an equivalent duration and depth of ischemia. However, despite the smaller infarct, aged mice have poorer outcomes and higher mortality compared to younger mice149–152. A few studies that utilized photochemically induced models of stroke have reported equivalent or larger infarcts in aged animals153,154. In addition to age, multiple factors including the species examined, the surgical method (reperfusion vs. permanent ischemia), and differences in collateral circulation (which can be strain-dependent in mice) influence the infarct volume after experimental stroke. However, in the majority of studies that utilized the transient MCAO model, aged mice consistently showed reduced edema and smaller infarcts yet have higher mortality and poorer neurological outcomes after stroke compared to their young controls149–151. Importantly, elderly stroke patients have a significantly different risk profile and comorbidities compared with younger patients.
Both intra- and inter-individual microbiota variations are drivers of stroke risk factors10. For instance, analysis of oral and fecal microbiota in participants of African-origin from multiple countries showed that oral Streptococcus, Prevotella, and Veillonella were enriched in participants with cerebrovascular risk factors including hypertension, hyperglycemia, and hypertriglyceridemia10. A study of 100 patients with atherosclerosis showed that plasma levels of tryptophan, indole, indole-3-propionate, and indole-3-aldehyde are negatively correlated with advancing atherosclerosis. Importantly, these specific indole metabolites are undetectable in GF mice155. In contrast, the kynurenine/tryptophan ratio was positively correlated with atherosclerosis156. This suggests that metabolic shunting of tryptophan metabolism away from the indole pathway (regulated by the microbiota) toward the kynurenine pathway (regulated by the host) may have detrimental effects on atherosclerosis, a major stroke risk factor. Table 3 provides a summary of major stroke risk factors (including atherosclerosis, small vessel disease, blood pressure, diabetes, obesity, and physical activity) and evidence for their link to the gut microbiota.
Table 3 -.
Major Stroke Risk Factors and Evidence for their Link to the Gut Microbiota
Risk | Link to Gut Microbiota |
---|---|
Atherosclerosis | • Patients with atherosclerosis showed that plasma levels of tryptophan, indole, indole-3-propionate, and indole-3-aldehyde are negatively correlated, but kynurenine/tryptophan ratio is positively correlated with advanced atherosclerosis (Cason et al. 2018). • Mediation analysis of fecal metagenomic data of 569 subclinical elderly in China showed that gut microbiota composition (Alistepes, Oligella, and Prevotella) explained 16.5% of beneficial effects of lifestyle (diet and exercise) on the pathogenesis of carotid atherosclerosis, assessed by intima-media thickness, peak systolic velocity, and end-diastolic velocity (Zhu et al. 2021) • Metagenome study on stools from 218 individuals with atherosclerotic cardiovascular disease (ACVD) showed increased abundance of Enterobacteriaceae and Streptococcus spp., higher potential for transport of simple sugars (e.g., phosphotransferase systems) and amino acids, and a lower potential for biosynthesis of most vitamins (Jie et al. 2017) • From Rotterdam Study, blood samples from 1111 participants showed that increase in concentration of 3-hydroxybutyrate is associated with increase in intracranial carotid artery calcification (ICAC), as a proxy of atherosclerosis volume (Vojinovic et al. 2018) • 132 differentially expressed genes (DEGs) were screened. Among the upregulated DEGs in CAS patients, FABP4 exhibited the highest increase, and FABP4 was positively associated with Acidaminococcus (Ji et al. 2021) |
Small Vessel Disease (SVD) | • A cross-sectional sub-analysis of 87 patients without dementia or a history of stroke showed that patients with high abundance of Bacteroides (>30%) were more likely to have cognitive decline, and some fecal metabolites (e.g., ammonia) were significantly higher in patients with higher total SVD scores compared with those with lower scores (Saji et al. 2021) |
Blood Pressure (BP) | • Fecal metagenome sequencing of 6953 Finns: the alpha (within‐sample) and beta (between‐sample) diversities were strongly related to BP indexes; positive associations between BP indexes and 27 distinct Firmicutes while negative associations with 19 distinct Lactobacillus spp. (Palmu et al. 2020) |
Diabetes Mellitus (DM) | • Treatment of high-fat mice with prebiotic (oligofructose) to increase Bifidobacterium led to improved glucose tolerance and reduced inflammation seen in diabetes and obesity (P D Cani et al. 2007) • Chronic high-fat diet increases LPS-containing gut microbiota and metabolic endotoxemia that can contribute to increased inflammatory tone, body weight gain, and initiation of diabetes in mice (Patrice D Cani et al. 2007) |
Obesity | • Diabetes and obesity are initiated and potentially transmissible via FMT • Humanized gnotobiotic mice fed high-fat, high-sugar diet shifted their microbiota structure and developed adiposity, and this effect was influenced by the dietary history of the human donors (Patrice D Cani et al. 2007) |
Physical Activity | • Exercise regulates microbiota (higher diversity, higher Firmicutes and Akkermansia, higher SCFAs) and exercise reduces depression/anxiety and increases BDNF (Monda et al. 2017) |
Stroke risk factors such as diabetes and obesity are transmissible via FMT (Table 3). Targeting microbiota to reduce metabolic dysfunction could be a clinical strategy to control metabolic risk factors for stroke. Dysfunction of the MGBA contributes to hypertension via metabolites, bacteria in the circulation, and autonomic dysregulation (recently reviewed here157). Accumulating evidence suggests that the gut microbiota can independently contribute to the development of important stroke risk factors. Therefore, these risk factors may be modulated or transmitted by the gut microbiota.
The Role Of The MGBA In Stroke Is Sex-Dependent.
Using an endothelin-1-induced middle cerebral artery occlusion (MCAO) model in rats of both sexes, results showed that males displayed greater mortality, worse sensory-motor deficit, and higher serum levels of proinflammatory cytokines IL-17A, MCP-1, and IL-5 as well as a more rapid stroke-induced gut permeability compared to females158. Analysis of subacute time points after MCAO in aged mice (14 days) demonstrated that male mice have higher mortality, higher rates of hemorrhagic transformation, a higher CD8+ T cell response in the blood and brain, and higher stroke-induced increases in gut permeability when compared to female mice even with similar infarct volumes at day 3159. These studies highlight the importance of sex as a biological variable in the MGBA after stroke160. Experiments utilizing cross-sex FMTs and sex-specific evaluation of the MGBA response after stroke are warranted.
Age-Related Frailty Is Associated With Dysbiosis And Poorer Stroke Outcomes.
A substantial proportion of elderly stroke patients are frail, which is linked to both cerebrovascular disease incidence and predicts shorter post-stroke survival161. Frailty is assessed by low grip strength, low energy, slowed walking speed, low physical activity, and unintentional weight loss162. A hallmark of frailty is a loss of microbiota diversity and taxonomic shifts, including increased Eubacterium dolichum and Eggerthella lenta, decreased Faecalibacterium prausnitzii, loss of SCFA-producers (e.g., butyrate-producers), decreased tryptophan metabolizers, and increased production of LPS and peptidoglycans163. Robust positive correlations exist between higher microbiota diversity and lower frailty, improved immune function, and enhanced cognitive performance164. For example, oral administration of Akkermansia sufficiently ameliorated the senescence-related phenotype in the intestines of aged mice and extended the health span, as evidenced by improvements in the frailty index and restoration of muscle atrophy131. These findings suggest that identifying key changes in microbial communities during aging can provide a rationale for developing bacteriotherapy to enhance healthy aging and post-stroke recovery.
The MGBA Plays A Role In Post-Stroke Complications.
Post-stroke intestinal ileus is a major contributor to stroke outcome, disability, and mortality. Clinically, ileus is characterized by abdominal distension and absent bowel sounds. Importantly, aging is a risk factor for developing post-stroke ileus. Analysis of Nationwide Inpatient Sample has shown that patients older than 75 are twice as likely to suffer from post-stroke ileus and related complications, compared to younger patients165. Post-stroke ileus can lead to bacterial overgrowth syndrome, increased intestinal barrier permeability, and nutrient malabsorption, which can exacerbate systemic inflammation. A placebo-control clinical trial of 55 patients with small intestinal bacterial overgrowth (SIBO) showed that weekly treatment with FMT capsules (prepared from healthy donors) for four weeks led to significantly increased bacterial diversity and markedly improved GI symptoms166. Mechanisms of post-stroke top-down signaling (likely via the ANS and circulatory factors) that contribute to ileus remain an important clinical question.
The Immune Response Is Different After Aged Stroke.
Abrupt reduction in blood flow at ischemic core leads to rapid cell death. The tissue at the periphery of this core (penumbra) remains at risk, yet salvageable167. The core injury initiates a cascade of inflammation and apoptosis triggered by glutamate excitotoxicity, aberrant calcium signaling, and oxidative stress, often exacerbated by reperfusion injury168. Relying on low affinity pattern recognition receptors (e.g., TLRs and RAGE), the CNS-resident, BAMs, and peripherally-sourced innate immune cells rapidly sense DAMPs, (e.g., ADP, HMGB1, and S100) generated immediately after vascular occlusion. The interruption in blood flow activates endothelial cells and initiates the coagulation cascade. BBB break down and brain-derived chemokine gradients are established, the complement system is activated, and adhesion and transmigration of blood-borne immune cells follows within minutes169,170.
Aging affects nearly all processes involved in the early response to stroke. For example, age-related increase in plasma levels of coagulation factors (e.g., fibrinogen, factors V, VII, VIII, IX, and XI) leads to a pro-coagulable state in the elderly and increases the risk of post-stroke complications171. Hemorrhagic transformation is more pronounced in aged versus young mice relative to infarct size, which is related to an earlier influx of neutrophils into the aged brain that produce metalloproteinases (MMPs) and leads to BBB disruption172. The subsequent complement activation and the recruitment of phagocytes and granulocytes (i.e., MG, mast cells, neutrophils, and macrophages) initiate the clearance of injured neuroglia173. Although necessary for debris clearance, persistent inflammation can lead to chronic tissue injury169. Aging alters the immunological response to ischemic stroke, and manipulating peripheral immunity by targeting the bone marrow or the microbiota can reverse this age-dependent response to stroke169,172.
MG Response After Stroke In Aged Animals:
Complete depletion of MG (using the dual CSF1R/c-Kit inhibitor PLX3397) worsens stroke outcomes174, suggesting that MG play a net beneficial role after stroke, likely by suppressing the post-stroke inflammation mediated by injured neuroglia. Aging affects the MG response to stroke101,172,175. Aged MG produce less TNF-α, but more ROS than young MG, and aged mice reconstituted with young bone marrow had reduced MG activation (lower FGF production and overall granularity) after stroke172. Additionally, post-stroke FMT from young healthy mice into aged mice reduced MG activation (decreased Iba1 and increased P2RY12)128. These results indicate that the age-dependent role of MG after stroke can be influenced by the manipulation of gut microbiota.
Innate Immune Response After Aged Stroke:
Post-stroke neutrophil infiltration into the brain is increased in aged animals172. Aged neutrophils show reduced phagocytosis but produce higher levels of ROS and MMP-9 after storke115,172. CNS BAMs, DCs and blood-derived monocytes are also among the early post-stroke responders. CCR2-mediated recruitment of monocyte-derived macrophages can be protective in the early phases of stroke by contributing to phagocytic activity and debris clearance; however, their continued presence contributes to chronic inflammation. Spleen and bone marrow are the primary sources of monocytes176 and contribute to systemic inflammation, including gut inflammation. In fact, upregulation of TLR4 expression on peripheral blood monocytes from acute stroke patients positively correlates with higher stroke severity and inflammatory injury177, implicating these monocytes in stroke-induced systemic inflammation. It is unclear whether age-related alterations in the microbiota can alter the post-stroke involvement of peripherally sourced neutrophils and monocytes.
DCs arrive early and are detected for several days after the onset of stroke178. Under the microbiota’s control, DCs contribute to inflammatory signaling by secreting IL-23, inducing the expression of IL-17 by γδ T cells, and promoting neutrophil infiltration179. The pre-stroke number of brain APCs is significantly higher in aged mice, and is correlated with specific age-related shifts in the microbiota (e.g., abundance of Akkermansia is negatively correlated with number of brain APCs)114. The post-stroke composition of infiltrating myeloid cells shows a larger monocytic contribution migrating into the young brain, but a larger neutrophilic contribution infiltrating into the aged brain 3 days after stroke172. The skull bone marrow houses significant populations of monocytes and neutrophils, with transcriptional signatures distinct from their blood-derived counterparts180. Importantly, the composition of myeloid populations in the skull bone marrow changes with aging with significantly higher neutrophils in the aged skull (Figure 4a, original unpublished data), which may explain the increased neutrophilic infiltration after stroke in aged compared to young mice.
Figure 4.
The composition of myeloid cell populations in the skull bone marrow changes with aging with significantly higher relative frequencies of neutrophils in aged skull (a). Skull bone marrow lymphocyte compartment contains significantly higher CD8+ T lymphocytes and activated CD11bhigh B lymphocytes in naïve aged mice when compared to young skull (b).
The Adaptive Immune Response After Stroke In Aging:
The subacute response begins 1 to 3 days after stroke. As a result of ischemic injury and local inflammation, critical interfaces including the BBB, meningeal lymphatics, and blood-CSF barriers become compromised169,181, allowing brain-derived antigens and cytokines to leak into the systemic circulation. With help from APCs (primarily DCs), the adaptive arm is engaged for an antigen-specific response and development of immune memory for the presented brain antigens. B and T lymphocytes undergo clonal expansions in lymphoid organs (e.g., spleen and cervical lymph nodes) and return to the circulation to mount a whole-body response. Increased circulating lymphocyte subsets (e.g., CD4+CD28− beyond 8%) have been linked to a higher risk of recurrence and mortality after stroke182. Increased IL-17+ T cells in the blood is associated with exacerbated cognitive deficits after stroke183,184, suggesting a contribution of antigenic activation of lymphocytes to chronic sequelae of stroke such as cognitive decline and autoimmunity. Indeed, autoreactive B and T cells are detected as early as four days after stroke, and circulating lymphocytes from stroke patients collected two weeks after their stroke show a more robust immunoreactivity to brain antigens (e.g., myelin) than those collected from controls185.
Pre-stroke bacterial colonization (by co-housing) from non-stroke SPF mice into GF mice was neuroprotective (reduced infarct volumes and increased IL-1β and TNF-α) in GF mice. This beneficial effect of microbiota was abolished in lymphocyte-deficient GF (GF Rag1−/−) mice, indicating that microbiota from healthy donors leads to lymphocyte-mediated neuroprotection after stroke186. Recent reports indicate that meningeal B cells, derived locally from the skull bone marrow, provide a constant supply of B cells exposed to brain antigens120. Additionally, we have found that skull bone marrow contains significantly higher CD8+ T lymphocytes and activated CD11bhigh B lymphocytes in naïve aged mice when compared to young skull (Figure 4b, original unpublished data). Blood-derived antigen-experienced B cells populate the meninges in aged mice, but not in young mice, in the absence of an acute CNS injury120.
Future studies are needed to determine whether these age-dependent heterogeneity of myeloid cells and lymphocytes contribute differently to the post-stroke response. Their proximity, temporal engagement, and cell-specific regulation by the gut microbiota after acute brain ischemia combined with the slower lymphatic clearance seen with aging187 may result in a prolonged exposure to novel CNS antigens that could contribute to increased incidence of autoimmunity and long-term cognitive decline seen in stroke patients188,189.
These findings indicate that aging alters the innate and adaptive immune response after stroke with an increased neutrophilic infiltration, an earlier involvement of peripherally-sourced APCs, and potentially different contribution of skull bone marrow-derived immune cells compared to immune cell sourced from the peripheral lymphoid organs (e.g, peripheral bone marrow, spleen, and thymus). It can be speculated that the age-related decrease in protective bacteria (e.g., Akkermansia or SCFA-producers) can lead to reduced intestinal barrier function. This then activates DCs to stimulate γδ T cells, increasing their migration to the brain and enhancing the production of IL-17, leading to the additional recruitment of neutrophils from the blood or skull bone marrow into the aged brain after stroke (Figure 3b). Future experiments are needed to clarify the role of age-related changes in the gut microbiota in cell-specific immune responses after stroke and in long term outcomes of stroke such as cognitive decline.
Aging Worsens Stroke-Induced Dysbiosis.
Post-stroke top-down signaling is mediated by: 1) the systemic immune response (e.g., monocytic and neutrophilic migration) to brain-originated signals, 2) activation of the HPA axis affecting the intestinal barrier function, and 3) activation of sympathetic and loss of parasympathetic drive disrupting ENS-mediated gut motility and GI secretions by EECs and Paneth cells. These top-down processes induce post-stroke dysbiosis, often super-imposed on the background of age-related dysbiosis and immunosenescence128,179,190. Post-stroke dysbiosis exacerbates systemic inflammation via bottom-up signaling, creating a vicious cycle of damage and ongoing inflammation.
Clinical Studies Of The Role Of MGBA After Stroke:
Table 4 provides an up-to-date summary of clinical studies focused on the role of MGBA in stroke. Studies have found a decrease in bacterial diversity in samples from stroke patients compared to healthy controls191,192. Yamashiro et al.193 found that increased Lactobacillus ruminis and reduced SCFAs (acetate and valerate) positively correlated with post-stroke inflammatory markers (elevated C-reactive protein and increased white blood cell counts) in a Japanese cohort of stroke patients. Xia et al.190 reported that Parabacteroides, Oscillospira, and Enterobacteriaceae were enriched in stroke patients, while Prevotella, Roseburia, and Faecalibacterium were reduced. Xu et al.194 showed that a higher abundance of Enterobacteriaceae was positively correlated with poor outcomes. A well-defined disease-specific microbial “fingerprint” for post-stroke dysbiosis requires large patient cohorts with appropriate controls for comorbidities, geography, diet, medication, race, sex, and age.
Table 4 -.
Summary of Clinical Studies Focused on the role of MGBA in Stroke
Country | Study type | Design (participants) | Microbiome Method & Intervention | Key Findings | Ref |
---|---|---|---|---|---|
China | Propective case-control study | 140 Stroke Patients (male:female = 95:45) vs. 92 controls (51:41), Asian (median age: 60) | Illumina MiSeq 16S rRNA amplicon sequencing on V4 region, blood and feces (stool within 72hrs of admission, no Abx info, stored @ -80C) | * Diversity: alpha diversity not reported. Significantly different beta diversity between stroke and control. * Phylum level: F:B ratio was higher in stroke group (small sample size, n=15) * Genus level: Decreased SCFA-producing bacteria: Roseburia, Bacteroides, Lachnospiraceae, Faecalibacterium, Blautia, and Anaerostipes in stroke group. Increased Lactobacillaceae, Akkermansia, Enterobacteriaceae, and Porphyromonadaceae in stroke group. |
(Tan et al., 2021) |
Netherlands | Prospective case-control study | 349 Stroke Patients (male:female = 194:155) vs. 51 controls (29:22), Caucasian (median age: 72) | Illumina MiSeq 16S rRNA amplicon sequencing on V3 region, blood and feces (rectal swab within 24hrs of adimission before Abx, stored @ -80C) | * Diversity: Increased alpha (within sample) diversity in stroke group but not in TIA. Significantly different beta (between-sample) diversity between stroke vs. control. * Phylum level: Reduced Firmicutes and Bacteriodetes while increased Proteobacteria in stroke group. No difference in F:B ratio. * Genus level: Increased Escherichia/Shigella, Peptoniphilus, Ezakiella, and Enterococcus in stroke group. Substantial decrease in obligate anaerobic genera including anaerostipes, Ruminococcus, and Subdoligranulum. * Metabolites: A loss of butyrate-producing bacteria in stroke group, which was also an independent predictor of higher risk of post-stroke infection. An increase of TMA producing-bacteria in stroke group. |
(Haak et al., 2021) |
China | Propective case-control study | 78 Stroke Patients (male:female = 49:29) vs. 98 controls (57:41), Asian (median age: 66) | Illumina MiSeq 16S rRNA amplicon sequencing on V4 region (stool, no timing/Abx info reported, stored @ -80C) | * Diversity: no significant difference in alpha or beta diversity. * Phylum level: Decreased Firmicutes and Bacteriodetes while increased Proteobacteria and Actinobacteria in stroke group. * Genus level: Decrease in six butyrate producing bacteria (Faecalibacterium, Subdoligranulum, Eubacterium recta, Roseburia, Lachnoclostridiu, and Butyricicoccus) in stroke group. Increase in lactic acid bacteria Lactobacillus and Lactococcus in stroke group. * butyrate-producing bacteria were negatively correlated with NIHSS while lactic acid bacteria were positively correlated with NIHSS. *Aging: Decreased Peptostreptococcaceae, Peptoclostridium, and Fusicatenibacter with age. |
(Li et al., 2020) |
China | Propective case-control study | 31 Stroke Patients (male:female = 22:9) vs. 9 controls (6:3), Asian (median age: 61) | Illumina MiSeq 16S rRNA amplicon sequencing on V4 region, blood and feces (stool within 48hrs of admission, preserved in anaerobic tube, patients on Abx excluded, stored @ -80C) | * Diversity: no significant difference in alpha or beta diversity. * Phylum level: not directly reported * Genus level: Decreased Blautia obeum in stroke group which was negatively correlated with WBC count. Increased Streptococcus infantis and Prevotela copri in stroke group and Streptococcus was positively correlated with creatinine and lipoprotein levels in stroke group. |
(Huang et al., 2019) |
China | Propective case-control study | 30 Stroke Patients (male:female = 21:9) vs. 30 controls (18:12), Asian (median age: 64) | Illumina MiSeq 16S rRNA amplicon sequencing on V1–2 region, blood and feces (stool within 48hrs of admission, patients on Abx excluded, stored @ -80C) | * Diversity: no significant difference in alpha or beta diversity. * Phylum level: no significant difference when comparing major phyla. * Genus level: Increased SCFA producers including Odoribacter, Akkermansia, Ruminococcaceae, and Victivallis. * Enterobacter was negatively correlated with NIHSS and mRS. |
(Li et al., 2019) |
China | Propective case-control study | 187 Stroke Patients (male:female = 78:26) vs. 160 controls (73:17), Asian (age: 18–80) | Illumina MiSeq 16S rRNA amplicon sequencing on V4 region, feces (stool within 48hrs of admission, patients on Abx/pre or pro-biotics, death within 7 days excluded, stored @ -80C) | * Diversity: Not directly reported. * Genus level: Increased Coprococcus, Fecalibacterium, Haemophilus, Knoellia, Lachnospira, Prevotella, Roseburia, Bradyrhizobiaceae, Clostridiaceae, Caulobacteraceae, and Erysipelotrichaeceae in stroke group. Decreased Bilophila, Butyricimonas, Oscillospira, Parabacteroides, Enterobacteriaceae, Rikenellaceae and Ruminococcaceae in stroke group. * A proposed index of dysbiosis (SDI) was proposed based on differences in relative abundance of enriched species between stroke and control group. The proposed SDI was tested against a training and validation cohort with promising predictive power for stroke severity. |
(Xia et al., 2019) |
US | Case-control perspective | 65 subjects of healthy and "non-healthy" (defined as cancer, CVD, pulmonary disease, diabetes, stroke or neurodegeneration | 16S (V1V3), fecal and saliva, | * Diversity: higher alpha-diversity in healthy aging compared to non-healthy aging. * Phylum level: Not directly reported. * Genus level: Akkermansia muciniphila and Erysipelotrichaceae UCG-003 were more abundant while Streptococcus was less abundant in healthy aging. * Metabolites: None reported. |
(Singh et al., 2019) |
US | Longitudinal | 36,429 subjects (all female), middle aged (40–59) and aged (>60) | Antibiotic use in different life-stages and durations | Longer duration of exposure to antibiotics in the middle and older adulthood was associated to an increased risk of cardiovascular disease. | (Heianza et al., 2019) |
US | Longitudinal | 3,359,653 US veterans with follow-up for ~7 year | Assessed constipation status and laxative use with all-cause mortality, coronary heart disease, and stroke | Constipation status and laxative use were independently associated with higher risk of all-cause mortality and ischemic stroke. | (Sumida et al., 2019) |
Switzerland | Prospective multicenter | 859 patients (>65 years) with acute venous thromboembolism (VTE) | Targeted mass spectrometry of TMAO by LC/MS | * Metabolites: TMAO shows a U-shaped risk association with mortality in elderly patients with acute VTE with the lowest risk found in medium plasma levels of TMAO. | (Reiner et al., 2019) |
China | case-controlled | 10 Stroke Patients (male:female = not reported) vs. 10 controls, (age: 53–82) | Illumina HiSeq2500 16S rRNA sequencing on V4 region, blood and feces | * Diversity: decreased alpha diversity (within sample) in stroke patients. No significant differences in beta diversity (between samples). * Phylum level: not directly reported. * Genus level: The most abundant genera in cases were Ruminococcus, Bacteroides, Prevotella, Parabacteroides, Dialister, Faecalibacterium, Megamonas, Roseburia, and Escherichia. A higher abundant of Gammaproteobacteria and a reduction in Bacteroidia were observed in cases. APOE had a positive correlation with Gammaproteobacteria while it was negatively correlated with Bacteroidia. |
(Wang et al., 2018) |
Germany | 78 Stroke patients for initial cohort and 593 patients for the validation cohort | Targeted mass spectrometry of TMAO by LC/MS and flow cytometry for blood monocytes | * Metabolites: that levels of proinflammatory Ly6C high monocytes were higher in mice fed with choline-rich diet to increase TMAO synthesis, but this increase in inflammatory monocytes was abolished if mice received antibiotic with choline-rich diet. | (Haghikia et al., 2018) | |
US | Case-control perspective | 100 Stroke patients (37% females) with advanced atherosclerosis vs. 22 age-/sex-matched controls (41% females) | Plasma metabolomics by tandem high performance liquid chromatography and mass spectrometry | * Metabolites: Plasma levels of tryptophan, indole, indole-3-propionate, and indole-3-aldehyde were negatively associated while kynurenine/tryptophan ratio was positively associated with advanced human atherosclerosis. | (Cason et al., 2018) |
Japan | fecal metabolomics | 41 Stroke Patients (male:female = 31:10) vs. 40 controls (24:16), Asian (age: 51–79) | 16S and 23S rRNA- targeted quantitative reverse transcription (qRT)-PCR on feces; high- performance liquid chromatography analyses on blood | * Stroke alters gut microbiota composition and reduces fecal SCFAs (acetate). * Gut microbiota were present in the blood of 7.5% controls and 4.9% cases. * Abundance of Lactobacillus ruminis was significantly higher in cases and it was positively correlated with IL-6 in serum. * The increased abundance of Atopobium cluster and L. ruminis as well as the reduced abundance of L. sakei subgroup were significantly associated with ischemic stroke. * Ischemic stroke was closely associated with low serum acetate and valerate. |
(Yamashiro et al., n.d.) |
England | A prospective exploratory observational study | 50 Stroke Patients and controls (male:female = 24:26), Caucasian (age: 65–86) | TOPO cloning and 16s rRNA gene sequencing, Salica swab (Oral cavity including buccal mucosa, tongue, gingiva and hard palate) | 103 bacterial phylotypes were found, of which 65% were Gram positive, 33% were Gram negative and 2% were Gram variable. 20 most common bacterial phylotypes included Streptococcus species (n = 14), Gram-negative Veillonella species (n = 3), Gram-positive Rothia mucilaginosa (n = 1), Gram-negative Treponema pedis (n = 1) and Gram-positive Lactobacillus fermentum (n=1). 30% of the acute stroke cases were diagnosed with at least one infection. | (Boaden et al., 2017) |
Australia | case-controlled | 36 Stroke Patients (male:female = 22:14) vs. 10 controls (all males), Caucasian (age: 53–71) | Illumina MiSeq 16S rRNA amplicon sequencing on V3-V4 region on blood, urine, lung tissues, and sputum | * More than 60% of the microbiota in the lungs were proven to originate from small intestines significantly (p = 0.01). * Stroke reduced the gut barrier permeability and caused intestinal dysfunction in order to allow the passage of microbiota into peripheral tissues. |
(Stanley et al., 2016) |
US | retrospective single-center study | 84 (44% men) IBD patients with thromboembolism | N/A | 83% of patients developed a venous thromboembolism. At the time of TE, 71% of patients were diagnosed with active IBD. | (Bollen et al., 2016) |
China | case-controlled, cross-sectional, observational | 322 AIS or TIA patients (male:female = 220:112) (median age of 61) vs. 231 controls (130:101), Asian (age: 45–80) | N/A | * Increased alpha-diversity, increased proteobacteria, decreased bacteriodes/prevotella/faecalibacterium, significantly wer blood TMAO concentrations in AIS and TIA groups compared to controls. * A higher abundance of Proteobacteria and a lower abundance of Bacteroides, Prevotella and Faecalibacterium were observed in cases. |
(Yin et al., 2015) |
China | retrospective single-center study | 18,392 patients with IBD vs. 73,568 non-IBD controls (all patients with prior stroke or diagnosed within one year of index date were excluded.) | N/A | The risk of ischemic stroke was 1.12 fold higher in IBD patients and signficantly increased with higher frequency of IBD exacerbations and hospitalization | (Huang et al., 2014) |
Canada | cross-sectional observational | 47 stroke patients (64% male, 71.8 median age) | N/A | Higher serum IL-17 and lower IL-10 levels were associated with worse cogivitve decline after stroke in patients with depressive symptoms but not those patients without depressive symptoms. | (Swardfager et al., 2014) |
US | Case-control perspective | 3903 patients (64% males) undergoing elective, non-urgent angiography, 3 years of follow-up | Targeted mass spectrometry of TMAO, choline, and betaine by LC/MS/MS | * Metabolites: Higher plasma levels of choline and betaine were associated with 1.9 and 1.4-fold increased risk of major adverse cardio/cerebro-vascular events (MACE) including stroke. Choline and betaine levels predicted future risk for MACE only when TMAO was elevated. | (Wang et al., 2014) |
Norway | serum metabolomics | 45 AIS patients versus 40 controls | N/A | Serum levels of tryptophan and tyrosine were lower in stroke group and reduced capacity for the synthesis of 5-hydroxytryptamine (serotonin) and catecholamines. | (Ormstad et al., 2013) |
US | Two perspective studies in parallel | 40 healthy adults with dietary challenge plus 4007 adults undergoing elective diagnostic cardiac catheterization | Targeted mass spectrometry of TMAO, choline, and betaine by LC/MS | * Metabolites: of plasma and urinary levels of TMAO after a dietary intake of TMAO precursors (e.g., phosphatidylcholine) in healthy participants before and after suppression of microbiota with antibiotics showed that TMAO production depends on metabolism by the microbiota, and higher TMAO levels are associated with increased risk of major adverse cardiovascular events (MACE). | (Tang et al., 2013) |
Belgium | serum metabolomics | 73 male/76 female patient samples at day 1, 3, and 7 after stroke onset | N/A | * Kynurenine/Trp ratio positively correlated with the NIHSS score, infarct volume, and poor outcome. * Kynurenine/Trp ratio at admission correlated with CRP levels, ESR, and NLR. |
(Brouns et al., 2010) |
China | blood flow cytometry | 49 AIS and 16 TIA patients 43% male) vs. 25 controls (55% male), samples at 24 hour after onset of symptoms | N/A | Number of TLR4+ monocytes was significantly increased in the blood in stroke patients and TLR4 expression correlated with stroke severity. | (Yang et al., 2008) |
US | blood flow cytometry | 56 men/50 women with or without stroke recurrent or death over one year follow-up | N/A | Elevated CD4+CD28- counts (>8%) in circulation was associated with increased (from 14.2% to 48.1%) risk of stroke recurrence and death. | (Nadareishvili et al., 2004) |
Pre-Clinical Studies Of The Role Of MGBA After Stroke:
Table 5 provides an up-to-date summary of pre-clinical studies focused on the role of MGBA in stroke, separated into studies that used young animal models versus those that used aged animal models of experimental stroke. In pre-clinical models, a complete absence of the microbiota in GF mice and antibiotic-treated mice leads to poorer stroke outcomes compared to SPF mice195, which suggest a net beneficial role of young microbiota. Moreover, pre-stroke bacterial colonization from non-stroke SPF mice into GF mice reduces infarct volumes, increases MG/macrophage counts, and increases brain cytokines (IL-1β and TNF-α) in GF mice, indicating that the pre-stroke microbiota from young healthy donors has the potential to play a neuroprotective role after stroke186. On the other hand, pre-stroke FMT from SPF mice with stroke increased infarct volume and worsened neurological deficits after stroke in GF mice196. Consistently, pre-stroke FMT from stroke patients into antibiotic-treated mice increased infarct volume and increased neurological deficits in recipient mice190. These results suggest that pre-stroke colonization with microbiota from healthy donors is neuroprotective, but colonization with a dysbiotic microbiota (as seen with aging, and in patients with prior stroke or other neurological disease) has deleterious effects on stroke outcomes. The effects of a prior stroke on the immune response to a new stroke is of significant clinical relevance and should be further investigated.
Table 5 -.
Summary of Preclinical Studies Focused on the role of the MGBA in Stroke (separated by age)
Age of Animals | Design | Key Findings | |
---|---|---|---|
Stroke Studies Performed in Young Animal Models | |||
Young | 104 patients with AIS and 90 healthy controls, microbiome sequencing, FMT from human patients (every 3 patients pooled) to antibiotic-treated young (6 weeks) mice before MCAO | • 18 genera significantly different between AIS and controls • Mice receiving FMT from stroke patients with higher index of dysbiosis showed worsened brain injury and elevated IL-17+ gd T cells in the gut. |
(Xia et al., 2019) |
Young | Young (7–10 weeks) mice subjected to MCAO | • Stroke-induced shift in mucosal microbiota composition included an increased abundance of Akkermansia muciniphila and an excessive abundance of clostridial species. • Predicted functional potential of the altered microbiota induced by stroke using PICRUSt revealed increases infectious processes, membrane transport and xenobiotic degradation pathways. |
(Stanley et al., 2018) |
Young | Young (10–12 weeks) mice subjected to permanent distal MCAO | • By comparing germfree mice with recolonized and conventional mice, it wash shown that bacterial colonization reduces stroke volumes. • Bacterial colonization increased cerebral expression of cytokines (Il-1b and Tnfa mRNA expressions) and increased microglia/macrophage cell counts (Iba1+ cells/unit area) in contrast to improved stroke outcome. • The microbiome-mediated brain protection after stroke was absent in lymphocyte-deficient mice, suggesting a neuroprotective role of microbiota in T cell priming after stroke. • Increased in overall T helper cell (CD4+) counts and polarized regulatory T cells (Foxp3+) and Th17 cells (RoRγt+) in the Peyer’s patches and in the spleens of germ free mice with conventional microbiota compared to germ free mice at five days after stroke. |
(Singh et al., 2018) |
Young | Young (10–14 weeks) mice subjected to MCAO | • Stroke induced cecal dysiobis (increased cecal peptococcaceae and Prevotellaceae) • Increased sympathetic tone (increased noradrenaline release and uptake) in the cecum • Reduced cecal mucoprotein production, and reduced number of goblet cells |
(Houlden et al., 2016) |
Young | Young (6 weeks) mice subjected to MCAO | • Antibiotic-induced alterations in the intestinal flora reduces ischemic brain injury in mice, an effect transmissible by fecal transplants. • Intestinal dysbiosis increased regulatory T cells and reduced IL-17+ γδ T cells, through altered dendritic cell activity. • Dysbiosis suppresses trafficking of effector T cells from the gut to the leptomeninges after stroke. • Interleukin-10 (IL-10) and IL-17 are required for the neuroprotection afforded by intestinal dysbiosis. |
(Benakis et al., 2016) |
Young | Young mice subjected to MCAO | • 24 hours after stroke, Peyer’s patches showed significant reduction of T and B cell counts after cerebral ischemia, while no differences in natural killer cells and macrophages were observed. | (Schulte-Herbrüggen et al., 2009) |
Not reported | Mice subjected to MCAO | • 14 days of probiotic (Bifidobacterium breve, Lactobacillus casei/bullgaricus/acidophilus) before MCAO reduced infarct size by 52% but did not improve neurological function 24 hour after MCAO. • Probiotics decreased the malondialdehyde content and the tumor necrosis factor-alpha levels in the ischemic brain tissue. |
(Akhoundzadeh et al., 2018) |
Not reported | Rats subjected to MCAO | • Significant time-dependent necrosis and shedding of the epithelium after stroke. • Significant increase of T lymphocytes in Peyer’s patches at 12 h and 24 h after stroke, while no differences in the number of B lymphocytes and the intraepithelial lymphocytes (IELs) were found. • Upregulation of CCL19 mRNA expression in the ileum was detected at 6 h after stroke. |
(Liu et al., 2017) |
Not reported | Mice subjected to permanent distal MCAO | • Reduced microbiota diversity and bacterial overgrowth of Bacteroidetes were identified as hallmarks of poststroke dysbiosis and associated with intestinal barrier dysfunction and reduced intestinal motility. • Recolonizing germ free mice with poststroke microbiota exacerbated lesion volume and functional deficits after experimental stroke in germ free mice compared with the recolonization with a normal control microbiota. • Number of IL-17+ and IFN-γ + CD4+ T cells were increased in Peyer’s patches of germ free mice recolonized with post-stroke-microbiota. • Number of CD11b+ monocytes/macrophages in the mucosal layer of the ileum was increased in germ free mice recolonized with the post-stroke microbiota compared with germ free mice receiving sham microbiota. |
(Singh et al., 2016) |
Not reported | Rats subjected to MCAO | • The serum ghrelin level was higher in the MCAO group. • The impelling force was lower in MCAO rats, reaching the lowest level at 24 hr post-MCAO. • Damage to the intestinal mucosa, including villus intestinalis, vacuolar degeneration of organelles, widened cell-cell junctions, and apoptotic cells could be found on microscopy. |
(Xu et al., 2012) |
Stroke Studies Performed in Aged Animal Models | |||
Aged | FMT from young (8–12 weeks) and aged (18–20 months) mice into young and aged recipients before MCAO | • The microbiota is altered after experimental stroke in young mice and resembles the biome of uninjured aged mice. • The ratio of Firmicutes to Bacteroidetes (F:B), two main bacterial phyla in gut microbiota, increased ∼9-fold (p < 0.001) compared to young. • Pre-stroke FMT from aged donors into young mic with MCAO increased mortality, decreased behavioral performance, and increased cytokine levels after MCAO. • Pre-stroke FMT from young donors into aged mic with MCAO increased survival and improved behavioral performance after MCAO. |
(Spychala et al., 2018) |
Aged | FMT from young (8–12 weeks) or aged (18–20 months) donors or oral gavage of SCFA-producers into aged male mice (18–20 months) after MCAO | • Post-stroke bacteriotherapy with four SCFA-producers (Bifidobacterium longum, Clostridium symbiosum, Faecalibacterium prausnitzii and Lactobacillus fermentum) alleviated post-stroke neurological deficits and inflammation, and elevated gut, brain and plasma SCFA concentrations in aged stroke mice. • At day 14 after stroke, aged stroke mice with young microbiome had less behavioral impairment, and reduced brain and gut inflammation. |
(Lee et al., 2020) |
Aged | Young (8–12 weeks) and aged (18–20 months) male mice subjected to MCAO | • While stroke induced gut permeability and bacterial translocation in both young and aged mice, only young mice were able to resolve infection. • Bacterial species seeding peripheral organs also differed between young (Escherichia) and aged (Enterobacter) mice. Consequently, aged mice developed a septic response marked by persistent and exacerbated hypothermia, weight loss, and immune dysfunction compared to young mice following stroke. • Higher gut permeability was observed in aged mice, and it led to high neurological deficits and higher mortality rate in aged mice after stroke. • Greater bacterial burden was observed in MLNs, spleen, liver, and lungs of aged stroke mice, correlated with higher rates of post-stroke sepsis. • Greater lymphocytic infiltration into the brain at 72 hours in aged compared to young mice. |
(Crapser et al., 2016) |
Importantly, aged mice not only have a baseline dysbiotic state, they are also more susceptible to stroke-induced dysbiosis as measured by increased gut permeability and sepsis due to bacterial translocation197. Aged mice receiving pre-stroke FMT from young donors have improved survival rates, reduced circulating cytokines (IL-6, TNF-α, Eotaxin, and CCL5), and improved behavioral outcomes after stroke127, even though infarct size was unaltered. This suggests that microbiota rejuvenation in older patients at risk for stroke could be a promising prophylactic therapy. With the exception of a few studies128, most preclinical studies have been performed in young mice and microbiota manipulations have been performed before stroke onset, both of which significantly reduce the translational value of the results (Table 5).
Microbiota-Based Treatments For Stroke
Probiotics And Prebiotics:
Probiotics (live bacteria) and prebiotics (selectively fermented ingredients that promote changes in the composition and activity of the microbiota) can be substitutes for clinical FMT. For instance, Panax notoginsenoside extract given before stroke is protective in rats by regulating GABA-b receptors via an increase in Bifidobacterium longum198. Pre-stroke administration of probiotics can also suppress the production of proinflammatory cytokines (TNF-α and IL-6), reduces hippocampal neuronal injury, and restores spatial memory behavior in a mouse hypoperfusion-model of stroke199. A recent meta-analysis showed that supplementation of enteral nutrition with probiotics in stroke patients is associated with decreased serum TNF-α, IL-6, and IL-10. Probiotics supplementation also reduced the incidence of post-stroke complications, including esophageal reflux, bloating, constipation, diarrhea, gastric retention, and gastrointestinal bleeding200.
Short-Chain Fatty Acids (SCFAs):
Dietary fibers and resistant starch are degraded via bacterial fermentation into acetate, propionate, butyrate, and other similar molecules (five carbons or less), known as SCFAs. SCFAs decrease intestinal inflammatory cytokines, regulate intestinal secretion, and enhance peristalsis. Although the host can produce some SCFAs (e.g., acetate201), large quantities of SCFAs are produced by the gut microbiota. SCFAs exert their effects in multiple ways, including binding G-protein coupled receptors (GPCRs), passive diffusion through the cell membrane, stimulation of histone acetyltransferases, inhibition of histone deacetylase 3 (HDAC3), stabilization of the hypoxia-inducible factor (HIF), and have vasoactive properties202,203. GPCRs for SCFAs (FFAR2/3) are expressed on epithelial, enteroendocrine204, and innate immune cells205, as well as on vagal afferent fibers206. SCFAs (butyrate) increased the production of AMPs and proinflammatory cytokines (IL-6 and TNF-α) by the intestinal epithelium, indicating a role for SCFAs in host anti-microbial defense207. SCFAs decreased the production of LPS-induced TNF-α, IL-1β and IL-6 by murine macrophages208. These findings suggest that the effects of SCFAs are cell- and pathway-specific. Further evidence is needed to determine whether physiologically relevant concentrations of SCFAs can reach the brain, given their short half-life (25 min – 3 hours)209.
SCFA-Producing Bacteria In Stroke:
Age-related loss of SCFA-producers has been linked to stroke risk factors, age-related cognitive decline, and stroke recovery128,202. Fecal metagenomic analysis of patients with hypertension shows a decline in SCFA utilization, and FMT from human donors with hypertension versus normotensive controls into GF mice show transmissibility of the hypertensive phenotype via the microbiota210. Mono-colonization of GF mice with butyrate-producing bacterium improves BBB integrity211. Our lab has shown that aged microbiota alone is sufficient to produce lower fecal SCFAs and led to enhanced cognitive deficits in GF mice, when compared to GF mice with young microbiota129/ This indicates that age-related dysbiosis (characterized by reduced SCFAs among other features) can contribute to stroke risk factors and cognitive decline in the absence of an acute ischemic stroke.
Emerging evidence indicates that microbiota composition can be therapeutically exploited to improve recovery even when initiated after stroke in aged mice either with young FMT or any targeted manipulation of the microbiota. Post-stroke bacteriotherapy with SCFA-producers (B. longum, C. symbiosum, F. prausnitzii, and L. fermentum) and inulin (a prebiotic substrate for SCFA producers) initiated three days after stroke, 1) increased brain and plasma levels of SCFAs, 2) increased expression of mucin-related genes and improved intestinal barrier function, 3) reduced the frequency of proinflammatory IL-17-producing γδ T cells in the brain (not in the intestines), and 4) improved stroke recovery (improved neurological deficits, motor function, grip strength, and depressive phenotypes) in aged mice128. These beneficial effects were independent of infarct size. Of significance, these effects on stroke outcomes were not related to the chronological age of the host, but instead were mediated by the age of the microbiota or its metabolites. Complementing this finding, a recent study showed that IL-17+ γδ T cells traffic from the intestinal LP to the leptomeninges and increase neuroinflammation after stroke179. The beneficial effects of SCFAs after stroke may be mediated by an increase in anti-inflammatory IL-10 and a decrease in gut-originated IL-17+ γδ T cells in the ischemic brain. These results indicate that post-stroke approaches targeting the microbiota composition and function can be a promising clinical strategy with an extended therapeutic window for the treatment of stroke.
Bile Acids (BAs) are synthesized from cholesterol in the liver and facilitate emulsification and adsorptions of lipids. BAs modify the composition of gut microbiota via AMPs and immune regulation. Conversely, gut microbiota convert primary BAs via bile salt hydrolysis and dehydroxylation to secondary BAs. BAs in the intestinal lumen signal to the CNS via direct pathways (systemic circulation) and indirect pathways, mediated through farnesoid X receptor (FXR) signaling and Takeda G protein-coupled receptor-GLP-1 (TGR5-GLP-1) signaling45. BAs disrupted BBB gap junction proteins (occludin) and increased BBB permeability212. A subset of EECs (L-cells) produce GLP-1 upon the activation of TGR5 by microbiota-regulated BAs. GLP-1 can be sensed by GLP-1 receptors expressed by afferent vagal fibers213, and is involved in glucose metabolism and energy homeostasis45. BAs represent a complex interplay of microbiota-host metabolism that influences the MGBA signaling.
BAs In Stroke:
BAs play a major role in cholesterol homeostasis, and are linked to development of atherosclerosis, a major risk factor for stroke. A recent study demonstrated that direct activation of CNS TGR5 signaling counteracts diet-induced obesity, whereas genetic downregulation of hypothalamic TGR5 promotes it214. BAs also regulate inflammation and metabolic disorders. For example, BAs inhibit NLRP3 inflammasome activation (a major host defense mechanism) via the TGR5-cAMP-PKA axis215. A single-center retrospective study of 777 AIS patients divided into four groups based on the quartile of the serum total BAs showed that increased admission serum BAs was associated with a reduced 3-month mortality after stroke216, suggesting protective effects of bile acids in ischemic stroke patients. Interestingly, patients with the highest serum BAs from the same study had the lowest WBC counts, hinting at potential anti-inflammatory effects of BAs. Another study showed that lower BA excretion in feces may be an independent risk factor for stroke217. It is challenging to reliably deduce mechanistic insights from high level correlative clinical data from different sample types (serum vs. feces). However, these retrospective studies clearly implicate the microbiota in BAs homeostasis, and BA-mediated regulation of multiple stroke risk factors. Future studies are necessary to determine the effects of age-related changes in the gut microbiota and stroke-induced dysbiosis on BAs metabolism in the context of stroke risk and recovery.
Bacteria That Regulate Tryptophan Metabolites:
Host metabolism of tryptophan is through the kynurenine pathway via tryptophan 2,3-dioxygenase and indoleamine 2,3-dioxygenase (TDO and IDO) as well as the 5HT pathway via tryptophan hydroxylase. Microbial metabolism of tryptophan is through the indole pathway by both tryptophanase-positive and tryptophanase-negative bacteria62. Microbiota-derived indole-based metabolites and host-derived kynurenine-based metabolites are physiologically relevant endogenous ligands for the AHR. For example, activation of AHR promotes differentiation of naïve T cells to anti-inflammatory Treg cells218, and activation of AHR by microbiota-dependent indole-based ligands reduces astrocyte-mediated neuroinflammation in EAE74. Tryptophan is also a precursor for 5HT synthesis, which can modulate the signaling between the host’s EECs and ANS, as described above219. Future studies are needed to examine the role of microbiota-dependent tryptophan-based ligands of AHR in post-stroke neuroinflammation.
Aging is associated with dysregulated amino acid metabolism, a contributor to immunosenescence and inflammaging101. Multiple amino acids, including glutamine, arginine, and tryptophan, have been linked to age-related remodeling of immunity101. Tryptophan is metabolized by both the host (kynurenine and serotonin pathways) and the microbiota (indole pathway). Aging is associated with increased IDO/TDO-mediated breakdown of tryptophan into kynurenine metabolites and leads to age-dependent elevated serum kynurenine. Metabolic shunting of tryptophan away from the indole pathway (regulated by the microbiota) toward the kynurenine pathway (regulated by the host) has been linked to the post-stroke inflammatory response220,221. Cuartero et al. showed that activation of AHR by kynurenine exacerbates acute brain damage following ischemia222. Pharmacological inhibition of the AHR after stroke reduced brain levels of inflammatory cytokines (IL-1β, IL-6, IFN-g, CXCL1, and S100b) and improved stroke outcomes222,223, suggesting that activation of AHR pathway by host-derived kynurenine metabolites plays a detrimental role in response to stroke. However, the role of AHR activation is highly ligand- and cell-specific74, and future studies are required to elucidate the effects of microbiota-derived (indole-based) versus host-derived (kynurenine-based) ligands of AHR after stroke.
Trimethylamine N-oxide (TMAO):
Diet has profound effects on the microbiota. For example, the production of TMAO by the microbiota can be increased by a high intake of dietary precursors of choline (L-carnitine and phosphatidylcholine), found in red meat and eggs224. Microbiota-derived TMAO has been linked to increased risk of cardiovascular events97,225–228. TMAO production involves the conversion of choline and L-carnitine by microbial lyases to TMA, an odorous gas. TMA reaches the liver via the portal vein, after which Flavin monooxygenases convert it to TMAO229. Clinical studies of plasma and urinary levels of TMAO after a dietary intake of TMAO precursors (e.g., phosphatidylcholine) in healthy participants before and after suppression of microbiota with antibiotics showed that TMAO production depends on metabolism by the microbiota228. Higher plasma levels of choline and betaine (a TMA-containing essential nutrient and its oxidation product) are associated with 1.9- and 1.4-fold increased risk of major adverse cardio/cerebrovascular events, including stroke230. Importantly, the detrimental effects of TMAO on stroke severity and outcomes can be transferred through FMT, provided that microbial lyases are present to convert choline to TMA231. Another study showed that levels of proinflammatory Ly6Chigh monocytes were higher in mice fed with choline-rich diet to increase TMAO synthesis, but this increase in inflammatory monocytes was abolished if mice received antibiotics with the choline-rich diet232. FMT studies have shown that stroke severity is transmissible and the TMAO pathway may be a mediator of this transmissibility231. Specifically, the microbial cutC gene (required for choline-to-TMA conversion) is sufficient to increase TMAO production and increase infarct size after stroke231. The role of TMAO in cerebrovascular disorders has been reviewed233. Improvement of diet may have a large effect on both the host and the microbiota.
Conclusion:
Our view of neurological disease has evolved from a single-organ brain centric view to a more integrated “whole-body” view. Stroke and neurodegenerative diseases induce systemic responses in which the gut microbiota play a key role. Mounting evidence supports the role of gut microbiota in age-dependent MGBA signaling after stroke. These observations encourage collaborations among historically segregated fields of microbiology, immunology, endocrinology, and neuroscience. In the clinical setting, differences in the microbiota composition are now being investigated to optimize treatment selection, to monitor treatment efficacy, and to enhance clinical prognosis234. Many essential questions remain to be answered, and limitations of preclinical models continue to cloud translational insights. Importantly, there remain significant differences between the common preclinical stroke models and typical stroke patients235. A typical mouse model utilizes a 10–12 week-old male (equivalent to a teenager) with no vascular risk factors. In contrast, a typical stroke patient is a 75-year-old with multiple vascular risk factors, taking multiple medications236. Appropriate animal models of aging and translational approaches are required to capitalize on the potential of manipulating the MGBA axis in aging and stroke. Understanding the role of this co-evolution of hosts and their microbiota provides new therapeutic insights to help patients suffering from neurodevelopmental, behavioral, neurodegenerative, and cerebrovascular diseases.
Figure 5.
Key contributors to differences between aged and young stroke and the role of age-related changes gut microbiota in response after stroke. A substantial proportion of elderly stroke patients are frail, which is linked to both cerebrovascular disease incidence and predicts shorter post-stroke survival. Post-stroke neutrophil infiltration into the brain and their ROS production are increased in aged animals after stroke. The pre-stroke relative frequency of brain APCs is significantly higher in aged mice. Skull bone marrow contains significantly higher neutrophils, CD8+ T lymphocytes, and activated CD11bhigh B lymphocytes in naïve aged mice compared to young skull. Major age-related changes in the immune and autonomic nervous system contribute to age-specific response after stroke. Aged microbiota is associated with reduced production of SCFAs and Trp metabolites, both of which are major regulators of immunity and physiological barriers (e.g., BBB and intestinal barrier). Post-stroke supplementation with SCFA-producers and inulin significantly reduces neuroinflammation and improves stroke outcomes. (Illustration credit: Ben Smith).
Financial Support
This work was supported by the NIH/National Institute of Neurological Disorders and Stroke (NINDS) Grant No. 1F31NS118984-01 (to PH), NIH/National Institute on Aging (NIA) and National Institute of Neurological Disorders and Stroke (NINDS): NIH/NINDS R01-NS103592 (Detrimental Effects of Age Related Dysbiosis to LDM and RMB), and NIH/ AG058463 (Dynamic non-neuronal interactions between the gut microbiota and the brain in aging to LDM and RMB).
Non-standard Abbreviations and Acronyms
- (MGBA)
The microbiota-gut-brain-axis
- (AD)
Alzheimer disease
- (PD)
Parkinson disease
- (MS)
Mass spectrometry
- (GLP-1)
Glucagon-Like Peptide 1
- (FMT)
Fecal Microbiota Transplant
- (SHIME)
Simulator of The Human Intestinal Microbial Ecosystem
- (GF)
Germ Free
- (GBB)
Gut-Blood Barrier
- (AMP)
Antimicrobial Peptides
- (M)
Microfold Cells
- (GALT)
Gut-Associated Lymphoid Tissues
- (LP)
Lamina Propria
- (PP)
Peyer’s Patches
- (APC)
Antigen Presenting Cell
- (EEC)
Enteroendocrine Cell
- 5HT
Serotonin (5-Hydroxy Tryptamine)
- (CCK)
Cholecystokinin
- (TLR)
Toll-Like Receptor
- (LPS)
Lipopolysaccharide
- (LN)
Lymph Node
- (BBB)
The Blood-Brain Barrier
- (CBF)
Cerebral Blood Flow
- (NO)
Nitric Oxide
- (NOS)
Nitric Oxide Synthase
- (MG)
Microglia
- (AHR)
Aryl Hydrocarbon Receptor
- (ACh)
Acetylcholine
- (ANS)
Autonomic Nervous System
- (NE)
Norepinephrine
- (NPY)
Neuropeptide Y
- (VIP)
Vasoactive Intestinal Peptide
- (ENS)
The enteric nervous system
- (VN)
Vagus nerve
- (EGC)
Enteric glial cell
- (HPA)
The hypothalamic-pituitary-adrenal axis
- (Aβ)
Amyloid-β
- (SCFA)
Short-chain fatty acid
- (GPCR)
G-protein coupled receptor
- (HDAC3)
Histone deacetylase 3
- (MCAO)
Middle cerebral artery occlusion
- (MMP)
Metalloproteinase
- (FXR)
Farnesoid x receptor
- (TGR5-GLP-1)
Takeda G protein-coupled receptor-GLP-1
- (TDO)
Tryptophan 2,3-dioxygenase
- (IDO)
Indoleamine 2,3-dioxygenase
- (TMAO)
Trimethylamine n-oxide
Footnotes
Conflict of Interest
None.
Some references do not include the enteric nervous system as part of the ANS.
Bibliography
- 1.Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017;17:219–232. doi: 10.1038/nri.2017.7 [DOI] [PubMed] [Google Scholar]
- 2.Ottman N, Smidt H, de Vos WM, Belzer C. The function of our microbiota: who is out there and what do they do? Front Cell Infect Microbiol. 2012;2:104. doi: 10.3389/fcimb.2012.00104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–14. doi: 10.1038/nature11234 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cryan JF, O’Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, et al. The Microbiota-Gut-Brain Axis. Physiol Rev. 2019;99:1877–2013. doi: 10.1152/physrev.00018.2018 [DOI] [PubMed] [Google Scholar]
- 5.Szabo G Gut-liver axis in alcoholic liver disease. Gastroenterology. 2015;148:30–6. doi: 10.1053/j.gastro.2014.10.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Jiao L, Kourkoumpetis T, Hutchinson D, Ajami NJ, Hoffman K, White DL, Graham DY, Hair C, Shah R, Kanwal F, et al. Spatial Characteristics of Colonic Mucosa-Associated Gut Microbiota in Humans. Microb Ecol. 2021;doi: 10.1007/s00248-021-01789-6 [DOI] [PubMed] [Google Scholar]
- 7.Vuik F, Dicksved J, Lam SY, Fuhler GM, van der Laan L, van de Winkel A, Konstantinov SR, Spaander M, Peppelenbosch MP, Engstrand L, Kuipers EJ. Composition of the mucosa-associated microbiota along the entire gastrointestinal tract of human individuals. United Eur Gastroenterol J. 2019;7:897–907. doi: 10.1177/2050640619852255 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Yeoh YK, Chen Z, Hui M, Wong MCS, Ho WCS, Chin ML, Ng SC, Chan FKL, Chan PKS. Impact of inter- and intra-individual variation, sample storage and sampling fraction on human stool microbial community profiles. PeerJ. 2019;7:e6172. doi: 10.7717/peerj.6172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kerimi A, Kraut NU, da Encarnacao JA, Williamson G. The gut microbiome drives inter- and intra-individual differences in metabolism of bioactive small molecules. Sci Rep. 2020;10:19590. doi: 10.1038/s41598-020-76558-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Fei N, Bernabé BP, Lie L, Baghdan D, Bedu-Addo K, Plange-Rhule J, Forrester TE, Lambert EV, Bovet P, Gottel N, et al. The human microbiota is associated with cardiometabolic risk across the epidemiologic transition. PLoS One. 2019;14:e0215262. doi: 10.1371/journal.pone.0215262 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.de Goffau MC, Lager S, Sovio U, Gaccioli F, Cook E, Peacock SJ, Parkhill J, Charnock-Jones DS, Smith GCS. Human placenta has no microbiome but can contain potential pathogens. Nature. 2019;572:329–334. doi: 10.1038/s41586-019-1451-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Shao Y, Forster SC, Tsaliki E, Vervier K, Strang A, Simpson N, Kumar N, Stares MD, Rodger A, Brocklehurst P, et al. Stunted microbiota and opportunistic pathogen colonization in caesarean-section birth. Nature. 2019;574:117–121. doi: 10.1038/s41586-019-1560-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, Kim SG, Li H, Gao Z, Mahana D, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell. 2014;158:705–721. doi: 10.1016/j.cell.2014.05.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.EBioMedicine. The maternal microbiome: another bridge linking mothers and infants. EBioMedicine. 2021;71:103602. doi: 10.1016/j.ebiom.2021.103602 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Morais LH, Schreiber HL, Mazmanian SK. The gut microbiota-brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2021;19:241–255. doi: 10.1038/s41579-020-00460-0 [DOI] [PubMed] [Google Scholar]
- 16.Cryan JF, O’Riordan KJ, Sandhu K, Peterson V, Dinan TG. The gut microbiome in neurological disorders. Lancet Neurol. 2020;19:179–194. doi: 10.1016/S1474-4422(19)30356-4 [DOI] [PubMed] [Google Scholar]
- 17.Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao J-Z, Abe F, Osawa R. Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol. 2016;16:90. doi: 10.1186/s12866-016-0708-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Willis AD. Rarefaction, Alpha Diversity, and Statistics. Front Microbiol. 2019;10:2407. doi: 10.3389/fmicb.2019.02407 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Finotello F, Mastrorilli E, Di Camillo B. Measuring the diversity of the human microbiota with targeted next-generation sequencing. Brief Bioinform. 2018;19:679–692. doi: 10.1093/bib/bbw119 [DOI] [PubMed] [Google Scholar]
- 20.Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102:11070–5. doi: 10.1073/pnas.0504978102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lozupone CA, Hamady M, Kelley ST, Knight R. Quantitative and qualitative beta diversity measures lead to different insights into factors that structure microbial communities. Appl Environ Microbiol. 2007;73:1576–85. doi: 10.1128/AEM.01996-06 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Langille MGI, Zaneveld J, Caporaso JG, McDonald D, Knights D, Reyes JA, Clemente JC, Burkepile DE, Vega Thurber RL, Knight R, et al. Predictive functional profiling of microbial communities using 16S rRNA marker gene sequences. Nat Biotechnol. 2013;31:814–21. doi: 10.1038/nbt.2676 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Yang JH, Bhargava P, McCloskey D, Mao N, Palsson BO, Collins JJ. Antibiotic-Induced Changes to the Host Metabolic Environment Inhibit Drug Efficacy and Alter Immune Function. Cell Host Microbe. 2017;22:757–765.e3. doi: 10.1016/j.chom.2017.10.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mahana D, Trent CM, Kurtz ZD, Bokulich NA, Battaglia T, Chung J, Müller CL, Li H, Bonneau RA, Blaser MJ. Antibiotic perturbation of the murine gut microbiome enhances the adiposity, insulin resistance, and liver disease associated with high-fat diet. Genome Med. 2016;8:48. doi: 10.1186/s13073-016-0297-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zarrinpar A, Chaix A, Xu ZZ, Chang MW, Marotz CA, Saghatelian A, Knight R, Panda S. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism. Nat Commun. 2018;9:2872. doi: 10.1038/s41467-018-05336-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Schwartz DJ, Langdon AE, Dantas G. Understanding the impact of antibiotic perturbation on the human microbiome. Genome Med. 2020;12:82. doi: 10.1186/s13073-020-00782-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ingber DE. Reverse Engineering Human Pathophysiology with Organs-on-Chips. Cell. 2016;164:1105–1109. doi: 10.1016/j.cell.2016.02.049 [DOI] [PubMed] [Google Scholar]
- 28.Van de Wiele T, Van den Abbeele P, Ossieur W, Possemiers S, Marzorati M. The Simulator of the Human Intestinal Microbial Ecosystem (SHIME®). 2015.
- 29.Petrof EO, Gloor GB, Vanner SJ, Weese SJ, Carter D, Daigneault MC, Brown EM, Schroeter K, Allen-Vercoe E. Stool substitute transplant therapy for the eradication of Clostridium difficile infection: “RePOOPulating” the gut. Microbiome. 2013;1:3. doi: 10.1186/2049-2618-1-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tan X, Johnson S. Fecal microbiota transplantation (FMT) for C. difficile infection, just say “No”. Anaerobe. 2019;60:102092. doi: 10.1016/j.anaerobe.2019.102092 [DOI] [PubMed] [Google Scholar]
- 31.Leslie JL, Huang S, Opp JS, Nagy MS, Kobayashi M, Young VB, Spence JR. Persistence and toxin production by Clostridium difficile within human intestinal organoids result in disruption of epithelial paracellular barrier function. Infect Immun. 2015;83:138–45. doi: 10.1128/IAI.02561-14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Spence JR, Mayhew CN, Rankin SA, Kuhar MF, Vallance JE, Tolle K, Hoskins EE, Kalinichenko VV, Wells SI, Zorn AM, et al. Directed differentiation of human pluripotent stem cells into intestinal tissue in vitro. Nature. 2011;470:105–9. doi: 10.1038/nature09691 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY, Ingber DE. Reconstituting organ-level lung functions on a chip. Science. 2010;328:1662–8. doi: 10.1126/science.1188302 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Benam KH, Novak R, Nawroth J, Hirano-Kobayashi M, Ferrante TC, Choe Y, Prantil-Baun R, Weaver JC, Bahinski A, Parker KK, Ingber DE. Matched-Comparative Modeling of Normal and Diseased Human Airway Responses Using a Microengineered Breathing Lung Chip. Cell Syst. 2016;3:456–466.e4. doi: 10.1016/j.cels.2016.10.003 [DOI] [PubMed] [Google Scholar]
- 35.Caruso R, Lo BC, Núñez G. Host-microbiota interactions in inflammatory bowel disease. Nat Rev Immunol. 2020;20:411–426. doi: 10.1038/s41577-019-0268-7 [DOI] [PubMed] [Google Scholar]
- 36.Nagpal J, Cryan JF. Microbiota-brain interactions: Moving toward mechanisms in model organisms. Neuron. 2021;109:3930–3953. doi: 10.1016/j.neuron.2021.09.036 [DOI] [PubMed] [Google Scholar]
- 37.Sadler R, Singh V, Benakis C, Garzetti D, Brea D, Stecher B, Anrather J, Liesz A. Microbiota differences between commercial breeders impacts the post-stroke immune response. Brain Behav Immun. 2017;66:23–30. doi: 10.1016/j.bbi.2017.03.011 [DOI] [PubMed] [Google Scholar]
- 38.Winek K, Meisel A, Dirnagl U. Gut microbiota impact on stroke outcome: Fad or fact? J Cereb Blood Flow Metab. 2016;36:891–8. doi: 10.1177/0271678X16636890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Deng Z, Luo XM, Liu J, Wang H. Quorum Sensing, Biofilm, and Intestinal Mucosal Barrier: Involvement the Role of Probiotic. Front Cell Infect Microbiol. 2020;10:538077. doi: 10.3389/fcimb.2020.538077 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Spadoni I, Zagato E, Bertocchi A, Paolinelli R, Hot E, Di Sabatino A, Caprioli F, Bottiglieri L, Oldani A, Viale G, et al. A gut-vascular barrier controls the systemic dissemination of bacteria. Science. 2015;350:830–4. doi: 10.1126/science.aad0135 [DOI] [PubMed] [Google Scholar]
- 41.Bonis V, Rossell C, Gehart H. The Intestinal Epithelium - Fluid Fate and Rigid Structure From Crypt Bottom to Villus Tip. Front cell Dev Biol. 2021;9:661931. doi: 10.3389/fcell.2021.661931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Macpherson AJ, Harris NL. Interactions between commensal intestinal bacteria and the immune system. Nat Rev Immunol. 2004;4:478–85. doi: 10.1038/nri1373 [DOI] [PubMed] [Google Scholar]
- 43.Bohórquez DV, Shahid RA, Erdmann A, Kreger AM, Wang Y, Calakos N, Wang F, Liddle RA. Neuroepithelial circuit formed by innervation of sensory enteroendocrine cells. J Clin Invest. 2015;125:782–6. doi: 10.1172/JCI78361 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Xue J, Askwith C, Javed NH, Cooke HJ. Autonomic nervous system and secretion across the intestinal mucosal surface. Auton Neurosci. 2007;133:55–63. doi: 10.1016/j.autneu.2007.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Mertens KL, Kalsbeek A, Soeters MR, Eggink HM. Bile Acid Signaling Pathways from the Enterohepatic Circulation to the Central Nervous System. Front Neurosci. 2017;11:617. doi: 10.3389/fnins.2017.00617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Mawe GM, Hoffman JM. Serotonin signalling in the gut--functions, dysfunctions and therapeutic targets. Nat Rev Gastroenterol Hepatol. 2013;10:473–86. doi: 10.1038/nrgastro.2013.105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Liu Q, Yang Q, Sun W, Vogel P, Heydorn W, Yu X-Q, Hu Z, Yu W, Jonas B, Pineda R, et al. Discovery and characterization of novel tryptophan hydroxylase inhibitors that selectively inhibit serotonin synthesis in the gastrointestinal tract. J Pharmacol Exp Ther. 2008;325:47–55. doi: 10.1124/jpet.107.132670 [DOI] [PubMed] [Google Scholar]
- 48.Cloutier N, Allaeys I, Marcoux G, Machlus KR, Mailhot B, Zufferey A, Levesque T, Becker Y, Tessandier N, Melki I, et al. Platelets release pathogenic serotonin and return to circulation after immune complex-mediated sequestration. Proc Natl Acad Sci U S A. 2018;115:E1550–E1559. doi: 10.1073/pnas.1720553115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.O’Connell PJ, Wang X, Leon-Ponte M, Griffiths C, Pingle SC, Ahern GP. A novel form of immune signaling revealed by transmission of the inflammatory mediator serotonin between dendritic cells and T cells. Blood. 2006;107:1010–7. doi: 10.1182/blood-2005-07-2903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.de las Casas-Engel M, Domínguez-Soto A, Sierra-Filardi E, Bragado R, Nieto C, Puig-Kroger A, Samaniego R, Loza M, Corcuera MT, Gómez-Aguado F, et al. Serotonin skews human macrophage polarization through HTR2B and HTR7. J Immunol. 2013;190:2301–10. doi: 10.4049/jimmunol.1201133 [DOI] [PubMed] [Google Scholar]
- 51.Bogunovic M, Davé SH, Tilstra JS, Chang DTW, Harpaz N, Xiong H, Mayer LF, Plevy SE. Enteroendocrine cells express functional Toll-like receptors. Am J Physiol Gastrointest Liver Physiol. 2007;292:G1770–83. doi: 10.1152/ajpgi.00249.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Osinski C, Le Gléau L, Poitou C, de Toro-Martin J, Genser L, Fradet M, Soula HA, Leturque A, Blugeon C, Jourdren L, et al. Type 2 diabetes is associated with impaired jejunal enteroendocrine GLP-1 cell lineage in human obesity. Int J Obes (Lond). 2021;45:170–183. doi: 10.1038/s41366-020-00694-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Legg LA, Tilney R, Hsieh C-F, Wu S, Lundström E, Rudberg A-S, Kutlubaev MA, Dennis M, Soleimani B, Barugh A, et al. Selective serotonin reuptake inhibitors (SSRIs) for stroke recovery. Cochrane database Syst Rev. 2019;2019. doi: 10.1002/14651858.CD009286.pub3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Oliver G, Kipnis J, Randolph GJ, Harvey NL. The Lymphatic Vasculature in the 21st Century: Novel Functional Roles in Homeostasis and Disease. Cell. 2020;182:270–296. doi: 10.1016/j.cell.2020.06.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sandrone S, Moreno-Zambrano D, Kipnis J, van Gijn J. A (delayed) history of the brain lymphatic system. Nat Med. 2019;25:538–540. doi: 10.1038/s41591-019-0417-3 [DOI] [PubMed] [Google Scholar]
- 56.Louveau A, Smirnov I, Keyes TJ, Eccles JD, Rouhani SJ, Peske JD, Derecki NC, Castle D, Mandell JW, Lee KS, et al. Structural and functional features of central nervous system lymphatic vessels. Nature. 2015;523:337–41. doi: 10.1038/nature14432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Absinta M, Ha S-K, Nair G, Sati P, Luciano NJ, Palisoc M, Louveau A, Zaghloul KA, Pittaluga S, Kipnis J, Reich DS. Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI. Elife. 2017;6. doi: 10.7554/eLife.29738 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Papadopoulos Z, Herz J, Kipnis J. Meningeal Lymphatics: From Anatomy to Central Nervous System Immune Surveillance. J Immunol. 2020;204:286–293. doi: 10.4049/jimmunol.1900838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Fitzpatrick Z, Frazer G, Ferro A, Clare S, Bouladoux N, Ferdinand J, Tuong ZK, Negro-Demontel ML, Kumar N, Suchanek O, et al. Gut-educated IgA plasma cells defend the meningeal venous sinuses. Nature. 2020;587:472–476. doi: 10.1038/s41586-020-2886-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Logsdon AF, Erickson MA, Rhea EM, Salameh TS, Banks WA. Gut reactions: How the blood-brain barrier connects the microbiome and the brain. Exp Biol Med (Maywood). 2018;243:159–165. doi: 10.1177/1535370217743766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Opp MR, George A, Ringgold KM, Hansen KM, Bullock KM, Banks WA. Sleep fragmentation and sepsis differentially impact blood-brain barrier integrity and transport of tumor necrosis factor-α in aging. Brain Behav Immun. 2015;50:259–265. doi: 10.1016/j.bbi.2015.07.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Rothhammer V, Mascanfroni ID, Bunse L, Takenaka MC, Kenison JE, Mayo L, Chao C-C, Patel B, Yan R, Blain M, et al. Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor. Nat Med. 2016;22:586–97. doi: 10.1038/nm.4106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Solár P, Zamani A, Kubíčková L, Dubový P, Joukal M. Choroid plexus and the blood-cerebrospinal fluid barrier in disease. Fluids Barriers CNS. 2020;17:35. doi: 10.1186/s12987-020-00196-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Faraci FM, Mayhan WG, Heistad DD. Effect of serotonin on blood flow to the choroid plexus. Brain Res. 1989;478:121–6. doi: 10.1016/0006-8993(89)91483-2 [DOI] [PubMed] [Google Scholar]
- 65.Ennis SR, Keep RF. Forebrain ischemia and the blood-cerebrospinal fluid barrier. Acta Neurochir Suppl. 2006;96:276–8. doi: 10.1007/3-211-30714-1_59 [DOI] [PubMed] [Google Scholar]
- 66.Lai Y, Liu C-W, Yang Y, Hsiao Y-C, Ru H, Lu K. High-coverage metabolomics uncovers microbiota-driven biochemical landscape of interorgan transport and gut-brain communication in mice. Nat Commun. 2021;12:6000. doi: 10.1038/s41467-021-26209-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Ma D, Wang AC, Parikh I, Green SJ, Hoffman JD, Chlipala G, Murphy MP, Sokola BS, Bauer B, Hartz AMS, Lin A-L. Ketogenic diet enhances neurovascular function with altered gut microbiome in young healthy mice. Sci Rep. 2018;8:6670. doi: 10.1038/s41598-018-25190-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Quirié A, Demougeot C, Bertrand N, Mossiat C, Garnier P, Marie C, Prigent-Tessier A. Effect of stroke on arginase expression and localization in the rat brain. Eur J Neurosci. 2013;37:1193–202. doi: 10.1111/ejn.12111 [DOI] [PubMed] [Google Scholar]
- 69.Kao CC, Cope JL, Hsu JW, Dwarkanath P, Karnes JM, Luna RA, Hollister EB, Thame MM, Kurpad AV, Jahoor F. The Microbiome, Intestinal Function, and Arginine Metabolism of Healthy Indian Women Are Different from Those of American and Jamaican Women. J Nutr. 2015;146:706–713. doi: 10.3945/jn.115.227579 [DOI] [PubMed] [Google Scholar]
- 70.Fouda AY, Eldahshan W, Narayanan SP, Caldwell RW, Caldwell RB. Arginase Pathway in Acute Retina and Brain Injury: Therapeutic Opportunities and Unexplored Avenues. Front Pharmacol. 2020;11:277. doi: 10.3389/fphar.2020.00277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Schulz O, Jaensson E, Persson EK, Liu X, Worbs T, Agace WW, Pabst O. Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions. J Exp Med. 2009;206:3101–14. doi: 10.1084/jem.20091925 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121–41. doi: 10.1016/j.cell.2014.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Rothhammer V, Borucki DM, Tjon EC, Takenaka MC, Chao C-C, Ardura-Fabregat A, de Lima KA, Gutiérrez-Vázquez C, Hewson P, Staszewski O, et al. Microglial control of astrocytes in response to microbial metabolites. Nature. 2018;557:724–728. doi: 10.1038/s41586-018-0119-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Rothhammer V, Quintana FJ. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol. 2019;19:184–197. doi: 10.1038/s41577-019-0125-8 [DOI] [PubMed] [Google Scholar]
- 75.Bechmann I, Kwidzinski E, Kovac AD, Simbürger E, Horvath T, Gimsa U, Dirnagl U, Priller J, Nitsch R. Turnover of rat brain perivascular cells. Exp Neurol. 2001;168:242–9. doi: 10.1006/exnr.2000.7618 [DOI] [PubMed] [Google Scholar]
- 76.Mrdjen D, Pavlovic A, Hartmann FJ, Schreiner B, Utz SG, Leung BP, Lelios I, Heppner FL, Kipnis J, Merkler D, et al. High-Dimensional Single-Cell Mapping of Central Nervous System Immune Cells Reveals Distinct Myeloid Subsets in Health, Aging, and Disease. Immunity. 2018;48:380–395.e6. doi: 10.1016/J.IMMUNI.2018.01.011 [DOI] [PubMed] [Google Scholar]
- 77.Williams JL, Jones SC, Page RB, Bryan RM. Vascular responses of choroid plexus during hypercapnia in rats. Am J Physiol. 1991;260:R1066–70. doi: 10.1152/ajpregu.1991.260.6.R1066 [DOI] [PubMed] [Google Scholar]
- 78.Faraci FM, Mayhan WG, Williams JK, Heistad DD. Effects of vasoactive stimuli on blood flow to choroid plexus. Am J Physiol. 1988;254:H286–91. doi: 10.1152/ajpheart.1988.254.2.H286 [DOI] [PubMed] [Google Scholar]
- 79.Schwartz M, Baruch K. The resolution of neuroinflammation in neurodegeneration: leukocyte recruitment via the choroid plexus. EMBO J. 2014;33:7–22. doi: 10.1002/embj.201386609 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Straub RH, Mayer M, Kreutz M, Leeb S, Schölmerich J, Falk W. Neurotransmitters of the sympathetic nerve terminal are powerful chemoattractants for monocytes. J Leukoc Biol. 2000;67:553–8. doi: 10.1002/jlb.67.4.553 [DOI] [PubMed] [Google Scholar]
- 81.Koopman FA, Stoof SP, Straub RH, Van Maanen MA, Vervoordeldonk MJ, Tak PP. Restoring the balance of the autonomic nervous system as an innovative approach to the treatment of rheumatoid arthritis. Mol Med. 17:937–48. doi: 10.2119/molmed.2011.00065 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Borgmann D, Ciglieri E, Biglari N, Brandt C, Cremer AL, Backes H, Tittgemeyer M, Wunderlich FT, Brüning JC, Fenselau H. Gut-brain communication by distinct sensory neurons differently controls feeding and glucose metabolism. Cell Metab. 2021;33:1466–1482.e7. doi: 10.1016/j.cmet.2021.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Fülling C, Dinan TG, Cryan JF. Gut Microbe to Brain Signaling: What Happens in Vagus…. Neuron. 2019;101:998–1002. doi: 10.1016/j.neuron.2019.02.008 [DOI] [PubMed] [Google Scholar]
- 84.Bellono NW, Bayrer JR, Leitch DB, Castro J, Zhang C, O’Donnell TA, Brierley SM, Ingraham HA, Julius D. Enterochromaffin Cells Are Gut Chemosensors that Couple to Sensory Neural Pathways. Cell. 2017;170:185–198.e16. doi: 10.1016/j.cell.2017.05.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Lv H, Zhao Y-H, Chen J-G, Wang D-Y, Chen H. Vagus Nerve Stimulation for Depression: A Systematic Review. Front Psychol. 2019;10:64. doi: 10.3389/fpsyg.2019.00064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Tang H, Li J, Zhou Q, Li S, Xie C, Niu L, Ma J, Li C. Vagus nerve stimulation alleviated cerebral ischemia and reperfusion injury in rats by inhibiting pyroptosis via α7 nicotinic acetylcholine receptor. Cell death Discov. 2022;8:54. doi: 10.1038/s41420-022-00852-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Dhawan S, De Palma G, Willemze RA, Hilbers FW, Verseijden C, Luyer MD, Nuding S, Wehkamp J, Souwer Y, de Jong EC, et al. Acetylcholine-producing T cells in the intestine regulate antimicrobial peptide expression and microbial diversity. Am J Physiol Gastrointest Liver Physiol. 2016;311:G920–G933. doi: 10.1152/ajpgi.00114.2016 [DOI] [PubMed] [Google Scholar]
- 88.Kimura I, Dohgu S, Takata F, Matsumoto J, Kawahara Y, Nishihira M, Sakada S, Saisho T, Yamauchi A, Kataoka Y. Activation of the α7 nicotinic acetylcholine receptor upregulates blood-brain barrier function through increased claudin-5 and occludin expression in rat brain endothelial cells. Neurosci Lett. 2019;694:9–13. doi: 10.1016/j.neulet.2018.11.022 [DOI] [PubMed] [Google Scholar]
- 89.Hansen MB. The enteric nervous system I: organisation and classification. Pharmacol Toxicol. 2003;92:105–13. doi: 10.1034/j.1600-0773.2003.t01-1-920301.x [DOI] [PubMed] [Google Scholar]
- 90.Savidge TC, Newman P, Pothoulakis C, Ruhl A, Neunlist M, Bourreille A, Hurst R, Sofroniew MV. Enteric glia regulate intestinal barrier function and inflammation via release of S-nitrosoglutathione. Gastroenterology. 2007;132:1344–58. doi: 10.1053/j.gastro.2007.01.051 [DOI] [PubMed] [Google Scholar]
- 91.McConalogue K, Furness JB. Gastrointestinal neurotransmitters. Baillieres Clin Endocrinol Metab. 1994;8:51–76. doi: 10.1016/s0950-351x(05)80226-5 [DOI] [PubMed] [Google Scholar]
- 92.Obata Y, Castaño Á, Boeing S, Bon-Frauches AC, Fung C, Fallesen T, de Agüero MG, Yilmaz B, Lopes R, Huseynova A, et al. Neuronal programming by microbiota regulates intestinal physiology. Nature. 2020;578:284–289. doi: 10.1038/s41586-020-1975-8 [DOI] [PubMed] [Google Scholar]
- 93.de Punder K, Pruimboom L. Stress induces endotoxemia and low-grade inflammation by increasing barrier permeability. Front Immunol. 2015;6:223. doi: 10.3389/fimmu.2015.00223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci. 2015;9:392. doi: 10.3389/fncel.2015.00392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Lo EH, Albers GW, Dichgans M, Donnan G, Esposito E, Foster R, Howells DW, Huang Y-G, Ji X, Klerman EB, et al. Circadian Biology and Stroke. Stroke. 2021;52:2180–2190. doi: 10.1161/STROKEAHA.120.031742 [DOI] [PubMed] [Google Scholar]
- 96.Mistry P, Reitz CJ, Khatua TN, Rasouli M, Oliphant K, Young ME, Allen-Vercoe E, Martino TA. Circadian influence on the microbiome improves heart failure outcomes. J Mol Cell Cardiol. 2020;149:54–72. doi: 10.1016/j.yjmcc.2020.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Parker A, Fonseca S, Carding SR. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes. 2019;1–23. doi: 10.1080/19490976.2019.1638722 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Muller PA, Schneeberger M, Matheis F, Wang P, Kerner Z, Ilanges A, Pellegrino K, Del Mármol J, Castro TBR, Furuichi M, et al. Microbiota modulate sympathetic neurons via a gut-brain circuit. Nature. 2020;583:441–446. doi: 10.1038/s41586-020-2474-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Moss C, Dhillo WS, Frost G, Hickson M. Gastrointestinal hormones: the regulation of appetite and the anorexia of ageing. J Hum Nutr Diet. 2012;25:3–15. doi: 10.1111/j.1365-277X.2011.01211.x [DOI] [PubMed] [Google Scholar]
- 100.Lejault P, Moruno-Manchon JF, Vemu SM, Honarpisheh P, Zhu L, Kim N, Urayama A, Monchaud D, McCullough LD, Tsvetkov AS. Regulation of autophagy by DNA G-quadruplexes. Autophagy. 2020;1–8. doi: 10.1080/15548627.2020.1769991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Conway J, Certo M, Lord JM, Mauro C, Duggal NA. Understanding the role of host metabolites in the induction of immunesenescence: future strategies for keeping the ageing population healthy. Br J Pharmacol. 2021;doi: 10.1111/bph.15671 [DOI] [PubMed] [Google Scholar]
- 102.Qi Y, Goel R, Kim S, Richards EM, Carter CS, Pepine CJ, Raizada MK, Buford TW. Intestinal Permeability Biomarker Zonulin is Elevated in Healthy Aging. J Am Med Dir Assoc. 2017;18:810.e1–810.e4. doi: 10.1016/j.jamda.2017.05.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Parrish AR. The impact of aging on epithelial barriers. Tissue barriers. 2017;5:e1343172. doi: 10.1080/21688370.2017.1343172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Singh H, Torralba MG, Moncera KJ, DiLello L, Petrini J, Nelson KE, Pieper R. Gastro-intestinal and oral microbiome signatures associated with healthy aging. GeroScience. 2019;41:907–921. doi: 10.1007/s11357-019-00098-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.van der Lugt B, van Beek AA, Aalvink S, Meijer B, Sovran B, Vermeij WP, Brandt RMC, de Vos WM, Savelkoul HFJ, Steegenga WT, Belzer C. Akkermansia muciniphila ameliorates the age-related decline in colonic mucus thickness and attenuates immune activation in accelerated aging Ercc1 -/Δ7 mice. Immun Ageing. 2019;16:6. doi: 10.1186/s12979-019-0145-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ahmadi S, Wang S, Nagpal R, Wang B, Jain S, Razazan A, Mishra SP, Zhu X, Wang Z, Kavanagh K, Yadav H. A human-origin probiotic cocktail ameliorates aging-related leaky gut and inflammation via modulating the microbiota/taurine/tight junction axis. JCI insight. 2020;5. doi: 10.1172/jci.insight.132055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.de Haan G, Lazare SS. Aging of hematopoietic stem cells. Blood. 2018;131:479–487. doi: 10.1182/blood-2017-06-746412 [DOI] [PubMed] [Google Scholar]
- 108.Zelante T, Iannitti RG, Cunha C, De Luca A, Giovannini G, Pieraccini G, Zecchi R, D’Angelo C, Massi-Benedetti C, Fallarino F, et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity. 2013;39:372–85. doi: 10.1016/j.immuni.2013.08.003 [DOI] [PubMed] [Google Scholar]
- 109.Lindemans CA, Calafiore M, Mertelsmann AM, O’Connor MH, Dudakov JA, Jenq RR, Velardi E, Young LF, Smith OM, Lawrence G, et al. Interleukin-22 promotes intestinal-stem-cell-mediated epithelial regeneration. Nature. 2015;528:560–564. doi: 10.1038/nature16460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ruiz-Ruiz S, Sanchez-Carrillo S, Ciordia S, Mena MC, Méndez-García C, Rojo D, Bargiela R, Zubeldia-Varela E, Martínez-Martínez M, Barbas C, et al. Functional microbiome deficits associated with ageing: Chronological age threshold. Aging Cell. 2020;19:e13063. doi: 10.1111/acel.13063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kaiko GE, Ryu SH, Koues OI, Collins PL, Solnica-Krezel L, Pearce EJ, Pearce EL, Oltz EM, Stappenbeck TS. The Colonic Crypt Protects Stem Cells from Microbiota-Derived Metabolites. Cell. 2016;165:1708–1720. doi: 10.1016/j.cell.2016.05.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Salazar N, Arboleya S, Fernández-Navarro T, de Los Reyes-Gavilán CG, Gonzalez S, Gueimonde M. Age-Associated Changes in Gut Microbiota and Dietary Components Related with the Immune System in Adulthood and Old Age: A Cross-Sectional Study. Nutrients. 2019;11. doi: 10.3390/nu11081765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Sovran B, Hugenholtz F, Elderman M, Van Beek AA, Graversen K, Huijskes M, Boekschoten MV, Savelkoul HFJ, De Vos P, Dekker J, Wells JM. Age-associated Impairment of the Mucus Barrier Function is Associated with Profound Changes in Microbiota and Immunity. Sci Rep. 2019;9:1437. doi: 10.1038/s41598-018-35228-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Honarpisheh P, Blixt FW, Blasco Conesa MP, Won W, D’Aigle J, Munshi Y, Hudobenko J, Weldon Furr J, Mobley A, Juneyoung Lee MS, et al. Peripherally-sourced myeloid antigen presenting cells increase with advanced aging. Brain Behav Immun. 2020;doi: 10.1016/j.bbi.2020.08.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Roy-O’Reilly MA, Ahnstedt H, Spychala MS, Munshi Y, Aronowski J, Sansing LH, McCullough LD. Aging exacerbates neutrophil pathogenicity in ischemic stroke. Aging (Albany NY). 2020;12:436–461. doi: 10.18632/aging.102632 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhang D, Chen G, Manwani D, Mortha A, Xu C, Faith JJ, Burk RD, Kunisaki Y, Jang J-E, Scheiermann C, et al. Neutrophil ageing is regulated by the microbiome. Nature. 2015;525:528–32. doi: 10.1038/nature15367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.McKenna RW, Washington LT, Aquino DB, Picker LJ, Kroft SH. Immunophenotypic analysis of hematogones (B-lymphocyte precursors) in 662 consecutive bone marrow specimens by 4-color flow cytometry. Blood. 2001;98:2498–507. doi: 10.1182/blood.v98.8.2498 [DOI] [PubMed] [Google Scholar]
- 118.Frasca D, Blomberg BB. Aging affects human B cell responses. J Clin Immunol. 2011;31:430–5. doi: 10.1007/s10875-010-9501-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Rubtsov AV, Rubtsova K, Fischer A, Meehan RT, Gillis JZ, Kappler JW, Marrack P. Toll-like receptor 7 (TLR7)-driven accumulation of a novel CD11c+ B-cell population is important for the development of autoimmunity. Blood. 2011;118:1305–15. doi: 10.1182/blood-2011-01-331462 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Brioschi S, Wang W-L, Peng V, Wang M, Shchukina I, Greenberg ZJ, Bando JK, Jaeger N, Czepielewski RS, Swain A, et al. Heterogeneity of meningeal B cells reveals a lymphopoietic niche at the CNS borders. Science. 2021;373. doi: 10.1126/science.abf9277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Krambs JR, Monlish DA, Gao F, Schuettpelz LG, Link DC. Microbiota Signals Suppress B Lymphopoiesis With Aging in Mice. Front Immunol. 2021;12:767267. doi: 10.3389/fimmu.2021.767267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Honarpisheh P, Reynolds CR, Conesa MPB, Manchon JFM, Putluri N, Bhattacharjee MB, Urayama A, McCullough LD, Ganesh BP. Dysregulated gut homeostasis observed prior to the accumulation of the brain amyloid-β in Tg2576 mice. Int J Mol Sci. 2020;21. doi: 10.3390/ijms21051711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Sun J, Xu J, Ling Y, Wang F, Gong T, Yang C, Ye S, Ye K, Wei D, Song Z, et al. Fecal microbiota transplantation alleviated Alzheimer’s disease-like pathogenesis in APP/PS1 transgenic mice. Transl Psychiatry. 2019;9:189. doi: 10.1038/s41398-019-0525-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Colombo AV, Sadler RK, Llovera G, Singh V, Roth S, Heindl S, Sebastian Monasor L, Verhoeven A, Peters F, Parhizkar S, et al. Microbiota-derived short chain fatty acids modulate microglia and promote Aβ plaque deposition. Elife. 2021;10. doi: 10.7554/eLife.59826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Rustenhoven J, Drieu A, Mamuladze T, de Lima KA, Dykstra T, Wall M, Papadopoulos Z, Kanamori M, Salvador AF, Baker W, et al. Functional characterization of the dural sinuses as a neuroimmune interface. Cell. 2021;184:1000–1016.e27. doi: 10.1016/j.cell.2020.12.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Louveau A, Herz J, Alme MN, Salvador AF, Dong MQ, Viar KE, Herod SG, Knopp J, Setliff JC, Lupi AL, et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat Neurosci. 2018;21:1380–1391. doi: 10.1038/s41593-018-0227-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Spychala MS, Venna VR, Jandzinski M, Doran SJ, Durgan DJ, Ganesh BP, Ajami NJ, Putluri N, Graf J, Bryan RM, McCullough LD. Age-related changes in the gut microbiota influence systemic inflammation and stroke outcome. Ann Neurol. 2018;84:23–36. doi: 10.1002/ana.25250 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lee J, D’Aigle J, Atadja L, Quaicoe V, Honarpisheh P, Ganesh BP, Hassan A, Graf J, Petrosino JF, Putluri N, et al. Gut Microbiota-Derived Short-Chain Fatty Acids Promote Post-Stroke Recovery in Aged Mice. Circ Res. 2020;CIRCRESAHA.119.316448. doi: 10.1161/CIRCRESAHA.119.316448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Lee J, Venna VR, Durgan DJ, Shi H, Hudobenko J, Putluri N, Petrosino J, McCullough LD, Bryan RM. Young versus aged microbiota transplants to germ-free mice: increased short-chain fatty acids and improved cognitive performance. Gut Microbes. 2020;12:1–14. doi: 10.1080/19490976.2020.1814107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Biagi E, Candela M, Fairweather-Tait S, Franceschi C, Brigidi P. Aging of the human metaorganism: the microbial counterpart. Age (Dordr). 2012;34:247–67. doi: 10.1007/s11357-011-9217-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Shin J, Noh J-R, Choe D, Lee N, Song Y, Cho S, Kang E-J, Go M-J, Ha SK, Chang D-H, et al. Ageing and rejuvenation models reveal changes in key microbial communities associated with healthy ageing. Microbiome. 2021;9:240. doi: 10.1186/s40168-021-01189-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Kim M-S, Kim Y, Choi HH, Kim W, Park S, Lee D-SD, Kim DK, Kim HJ, Choi HH, Hyun D-W, et al. Transfer of a healthy microbiota reduces amyloid and tau pathology in an Alzheimer’s disease animal model. Gut. 2020;69:283–294. doi: 10.1136/gutjnl-2018-317431 [DOI] [PubMed] [Google Scholar]
- 133.Sumida K, Molnar MZ, Potukuchi PK, Thomas F, Lu JL, Yamagata K, Kalantar-Zadeh K, Kovesdy CP. Constipation and risk of death and cardiovascular events. Atherosclerosis. 2019;281:114–120. doi: 10.1016/j.atherosclerosis.2018.12.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.O’Mahony D, O’Leary P, Quigley EMM. Aging and intestinal motility: a review of factors that affect intestinal motility in the aged. Drugs Aging. 2002;19:515–27. doi: 10.2165/00002512-200219070-00005 [DOI] [PubMed] [Google Scholar]
- 135.Phillips RJ, Powley TL. Innervation of the gastrointestinal tract: patterns of aging. Auton Neurosci. 2007;136:1–19. doi: 10.1016/j.autneu.2007.04.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Kim S-E. Colonic Slow Transit Can Cause Changes in the Gut Environment Observed in the Elderly. J Neurogastroenterol Motil. 2017;23:3–4. doi: 10.5056/jnm16215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Hotta H, Uchida S. Aging of the autonomic nervous system and possible improvements in autonomic activity using somatic afferent stimulation. Geriatr Gerontol Int. 2010;10 Suppl 1:S127–36. doi: 10.1111/j.1447-0594.2010.00592.x [DOI] [PubMed] [Google Scholar]
- 138.Toral M, Robles-Vera I, de la Visitación N, Romero M, Yang T, Sánchez M, Gómez-Guzmán M, Jiménez R, Raizada MK, Duarte J. Critical Role of the Interaction Gut Microbiota - Sympathetic Nervous System in the Regulation of Blood Pressure. Front Physiol. 2019;10:231. doi: 10.3389/fphys.2019.00231 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Saffrey MJ. Cellular changes in the enteric nervous system during ageing. Dev Biol. 2013;382:344–55. doi: 10.1016/j.ydbio.2013.03.015 [DOI] [PubMed] [Google Scholar]
- 140.Peck CJ, Samsuria SD, Harrington AM, King SK, Hutson JM, Southwell BR. Fall in density, but not number of myenteric neurons and circular muscle nerve fibres in guinea-pig colon with ageing. Neurogastroenterol Motil. 2009;21:1075–e90. doi: 10.1111/j.1365-2982.2009.01349.x [DOI] [PubMed] [Google Scholar]
- 141.Jurk D, Wang C, Miwa S, Maddick M, Korolchuk V, Tsolou A, Gonos ES, Thrasivoulou C, Saffrey MJ, Cameron K, von Zglinicki T. Postmitotic neurons develop a p21-dependent senescence-like phenotype driven by a DNA damage response. Aging Cell. 2012;11:996–1004. doi: 10.1111/j.1474-9726.2012.00870.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Phillips RJ, Kieffer EJ, Powley TL. Loss of glia and neurons in the myenteric plexus of the aged Fischer 344 rat. Anat Embryol (Berl). 2004;209:19–30. doi: 10.1007/s00429-004-0426-x [DOI] [PubMed] [Google Scholar]
- 143.Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng S, Chiuve SE, Cushman M, Delling FN, Deo R, et al. Heart Disease and Stroke Statistics-2018 Update: A Report From the American Heart Association. Circulation. 2018;137:e67–e492. doi: 10.1161/CIR.0000000000000558 [DOI] [PubMed] [Google Scholar]
- 144.Powers WJ, Rabinstein AA, Ackerson T, Adeoye OM, Bambakidis NC, Becker K, Biller J, Brown M, Demaerschalk BM, Hoh B, et al. Guidelines for the Early Management of Patients With Acute Ischemic Stroke: 2019 Update to the 2018 Guidelines for the Early Management of Acute Ischemic Stroke: A Guideline for Healthcare Professionals From the American Heart Association/American Stroke. Stroke. 2019;50:e344–e418. doi: 10.1161/STR.0000000000000211 [DOI] [PubMed] [Google Scholar]
- 145.Rinaldo L, Rabinstein AA, Cloft H, Knudsen JM, Castilla LR, Brinjikji W. Racial and Ethnic Disparities in the Utilization of Thrombectomy for Acute Stroke. Stroke. 2019;50:2428–2432. doi: 10.1161/STROKEAHA.118.024651 [DOI] [PubMed] [Google Scholar]
- 146.GBD 2013 DALYs and HALE Collaborators, Murray CJL, Barber RM, Foreman KJ, Abbasoglu Ozgoren A, Abd-Allah F, Abera SF, Aboyans V, Abraham JP, Abubakar I, et al. Global, regional, and national disability-adjusted life years (DALYs) for 306 diseases and injuries and healthy life expectancy (HALE) for 188 countries, 1990–2013: quantifying the epidemiological transition. Lancet (London, England). 2015;386:2145–91. doi: 10.1016/S0140-6736(15)61340-X [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Popović N, Stefanović-Budimkić M, Mitrović N, Urošević A, Milošević B, Pelemiš M, Jevtović D, Beslać-Bumbaširević L, Jovanović D. The frequency of poststroke infections and their impact on early stroke outcome. J Stroke Cerebrovasc Dis. 2013;22:424–9. doi: 10.1016/j.jstrokecerebrovasdis.2013.03.003 [DOI] [PubMed] [Google Scholar]
- 148.Camara-Lemarroy CR, Ibarra-Yruegas BE, Gongora-Rivera F. Gastrointestinal complications after ischemic stroke. J Neurol Sci. 2014;346:20–5. doi: 10.1016/j.jns.2014.08.027 [DOI] [PubMed] [Google Scholar]
- 149.Liu F, McCullough LD. Middle cerebral artery occlusion model in rodents: methods and potential pitfalls. J Biomed Biotechnol. 2011;2011:464701. doi: 10.1155/2011/464701 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Liu F, Yuan R, Benashski SE, McCullough LD. Changes in experimental stroke outcome across the life span. J Cereb Blood Flow Metab. 2009;29:792–802. doi: 10.1038/jcbfm.2009.5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Shapira S, Sapir M, Wengier A, Grauer E, Kadar T. Aging has a complex effect on a rat model of ischemic stroke. Brain Res. 2002;925:148–58. doi: 10.1016/s0006-8993(01)03270-x [DOI] [PubMed] [Google Scholar]
- 152.Sieber MW, Claus RA, Witte OW, Frahm C. Attenuated inflammatory response in aged mice brains following stroke. PLoS One. 2011;6:e26288. doi: 10.1371/journal.pone.0026288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Wang R-Y, Wang PS-G, Yang Y-R. Effect of age in rats following middle cerebral artery occlusion. Gerontology. 49:27–32. doi: 10.1159/000066505 [DOI] [PubMed] [Google Scholar]
- 154.Kharlamov A, Kharlamov E, Armstrong DM. Age-dependent increase in infarct volume following photochemically induced cerebral infarction: putative role of astroglia. J Gerontol A Biol Sci Med Sci. 2000;55:B135–41; discussion B142–3. doi: 10.1093/gerona/55.3.b135 [DOI] [PubMed] [Google Scholar]
- 155.Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106:3698–3703. doi: 10.1073/pnas.0812874106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Cason CA, Dolan KT, Sharma G, Tao M, Kulkarni R, Helenowski IB, Doane BM, Avram MJ, McDermott MM, Chang EB, et al. Plasma microbiome-modulated indole- and phenyl-derived metabolites associate with advanced atherosclerosis and postoperative outcomes. J Vasc Surg. 2018;68:1552–1562.e7. doi: 10.1016/j.jvs.2017.09.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Richards EM, Li J, Stevens BR, Pepine CJ, Raizada MK. Gut Microbiome and Neuroinflammation in Hypertension. Circ Res. 2022;130:401–417. doi: 10.1161/CIRCRESAHA.121.319816 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.El-Hakim Y, Mani KK, Eldouh A, Pandey S, Grimaldo MT, Dabney A, Pilla R, Sohrabji F. Sex differences in stroke outcome correspond to rapid and severe changes in gut permeability in adult Sprague-Dawley rats. Biol Sex Differ. 2021;12:14. doi: 10.1186/s13293-020-00352-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Ahnstedt H, Patrizz A, Chauhan A, Roy-O’Reilly M, Furr JW, Spychala MS, D’Aigle J, Blixt FW, Zhu L, Bravo Alegria J, McCullough LD. Sex differences in T cell immune responses, gut permeability and outcome after ischemic stroke in aged mice. Brain Behav Immun. 2020;87:556–567. doi: 10.1016/j.bbi.2020.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Honarpisheh P, McCullough LD. Sex as a biological variable in the pathology and pharmacology of neurodegenerative and neurovascular diseases. Br J Pharmacol. 2019;176:4173–4192. doi: 10.1111/bph.14675 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Palmer K, Vetrano DL, Padua L, Romano V, Rivoiro C, Scelfo B, Marengoni A, Bernabei R, Onder G. Frailty Syndromes in Persons With Cerebrovascular Disease: A Systematic Review and Meta-Analysis. Front Neurol. 2019;10:1255. doi: 10.3389/fneur.2019.01255 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Fried LP, Tangen CM, Walston J, Newman AB, Hirsch C, Gottdiener J, Seeman T, Tracy R, Kop WJ, Burke G, et al. Frailty in older adults: evidence for a phenotype. J Gerontol A Biol Sci Med Sci. 2001;56:M146–56. doi: 10.1093/gerona/56.3.m146 [DOI] [PubMed] [Google Scholar]
- 163.Jackson MA, Jackson M, Jeffery IB, Beaumont M, Bell JT, Clark AG, Ley RE, O’Toole PW, Spector TD, Steves CJ. Signatures of early frailty in the gut microbiota. Genome Med. 2016;8:8. doi: 10.1186/s13073-016-0262-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Claesson MJ, Jeffery IB, Conde S, Power SE, O’Connor EM, Cusack S, Harris HMB, Coakley M, Lakshminarayanan B, O’Sullivan O, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488:178–84. doi: 10.1038/nature11319 [DOI] [PubMed] [Google Scholar]
- 165.Rumalla K, Kumar AS, Mittal MK. Gastrointestinal Bowel Obstruction in Acute Ischemic Stroke: Incidence, Risk Factors, and Outcomes in a U.S. Nationwide Analysis of 3,998,667 Hospitalizations. J Stroke Cerebrovasc Dis. 2017;26:2093–2101. doi: 10.1016/j.jstrokecerebrovasdis.2017.04.028 [DOI] [PubMed] [Google Scholar]
- 166.Xu F, Li N, Wang C, Xing H, Chen D, Wei Y. Clinical efficacy of fecal microbiota transplantation for patients with small intestinal bacterial overgrowth: a randomized, placebo-controlled clinic study. BMC Gastroenterol. 2021;21:54. doi: 10.1186/s12876-021-01630-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Astrup J, Siesjö BK, Symon L. Thresholds in cerebral ischemia - the ischemic penumbra. Stroke. 12:723–5. doi: 10.1161/01.str.12.6.723 [DOI] [PubMed] [Google Scholar]
- 168.Spychala MS, Honarpisheh P, McCullough LD. Sex differences in neuroinflammation and neuroprotection in ischemic stroke. J Neurosci Res. 2017;95:462–471. doi: 10.1002/jnr.23962 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Iadecola C, Buckwalter MS, Anrather J. Immune responses to stroke: Mechanisms, modulation, and therapeutic potential. J Clin Invest. 2020;130:2777–2788. doi: 10.1172/JCI135530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Yilmaz G, Granger DN. Leukocyte recruitment and ischemic brain injury. Neuromolecular Med. 2010;12:193–204. doi: 10.1007/s12017-009-8074-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Tschan SL, Bolliger D. Coagulation and Aging: Implications for the Anesthesiologist. Curr Anesthesiol Rep. 2021;doi: 10.1007/s40140-021-00498-7 [DOI] [Google Scholar]
- 172.Ritzel RM, Lai Y-J, Crapser JD, Patel AR, Schrecengost A, Grenier JM, Mancini NS, Patrizz A, Jellison ER, Morales-Scheihing D, et al. Aging alters the immunological response to ischemic stroke. Acta Neuropathol. 2018;136:89–110. doi: 10.1007/s00401-018-1859-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Schilling M, Besselmann M, Leonhard C, Mueller M, Ringelstein EB, Kiefer R. Microglial activation precedes and predominates over macrophage infiltration in transient focal cerebral ischemia: a study in green fluorescent protein transgenic bone marrow chimeric mice. Exp Neurol. 2003;183:25–33. doi: 10.1016/s0014-4886(03)00082-7 [DOI] [PubMed] [Google Scholar]
- 174.Jin W-N, Shi SX-Y, Li Z, Li M, Wood K, Gonzales RJ, Liu Q. Depletion of microglia exacerbates postischemic inflammation and brain injury. J Cereb Blood Flow Metab. 2017;37:2224–2236. doi: 10.1177/0271678X17694185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Honarpisheh P, Lee J, Banerjee A, Blasco-Conesa MP, Honarpisheh P, D’Aigle J, Mamun AA, Ritzel RM, Chauhan A, Ganesh BP, McCullough LD. Potential caveats of putative microglia-specific markers for assessment of age-related cerebrovascular neuroinflammation. J Neuroinflammation. 2020;17:366. doi: 10.1186/s12974-020-02019-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Chauhan A, Al Mamun A, Spiegel G, Harris N, Zhu L, McCullough LD. Splenectomy protects aged mice from injury after experimental stroke. Neurobiol Aging. 2018;61:102–111. doi: 10.1016/j.neurobiolaging.2017.09.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Yang Q, Li J, Lu F, Wen A, Xiang J, Zhang L, Huang Z, Wang J. Upregulated expression of toll-like receptor 4 in monocytes correlates with severity of acute cerebral infarction. J Cereb Blood Flow Metab. 2008;28:1588–96. doi: 10.1038/jcbfm.2008.50 [DOI] [PubMed] [Google Scholar]
- 178.Felger JC, Abe T, Kaunzner UW, Gottfried-Blackmore A, Gal-Toth J, McEwen BS, Iadecola C, Bulloch K. Brain dendritic cells in ischemic stroke: time course, activation state, and origin. Brain Behav Immun. 2010;24:724–37. doi: 10.1016/j.bbi.2009.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Benakis C, Brea D, Caballero S, Faraco G, Moore J, Murphy M, Sita G, Racchumi G, Ling L, Pamer EG, et al. Commensal microbiota affects ischemic stroke outcome by regulating intestinal γδ T cells. Nat Med. 2016;22:516–23. doi: 10.1038/nm.4068 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Cugurra A, Mamuladze T, Rustenhoven J, Dykstra T, Beroshvili G, Greenberg ZJ, Baker W, Papadopoulos Z, Drieu A, Blackburn S, et al. Skull and vertebral bone marrow are myeloid cell reservoirs for the meninges and CNS parenchyma. Science (80- ). 2021;373. doi: 10.1126/science.abf7844 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Bernardo-Castro S, Sousa JA, Brás A, Cecília C, Rodrigues B, Almendra L, Machado C, Santo G, Silva F, Ferreira L, et al. Pathophysiology of Blood-Brain Barrier Permeability Throughout the Different Stages of Ischemic Stroke and Its Implication on Hemorrhagic Transformation and Recovery. Front Neurol. 2020;11:594672. doi: 10.3389/fneur.2020.594672 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Nadareishvili ZG, Li H, Wright V, Maric D, Warach S, Hallenbeck JM, Dambrosia J, Barker JL, Baird AE. Elevated pro-inflammatory CD4+CD28− lymphocytes and stroke recurrence and death. Neurology. 2004;63:1446–51. doi: 10.1212/01.wnl.0000142260.61443.7c [DOI] [PubMed] [Google Scholar]
- 183.Yin Y, Li G. Hypoxia induces T Helper 17 cell upregulation in cultured peripheral blood mononuclear cells from chronic stage patients of severe cerebral infarction. Microbiol Immunol. 2011;55:130–4. doi: 10.1111/j.1348-0421.2010.00301.x [DOI] [PubMed] [Google Scholar]
- 184.Swardfager W, Herrmann N, Andreazza AC, Swartz RH, Khan MM, Black SE, Lanctôt KL. Poststroke neuropsychiatric symptoms: relationships with IL-17 and oxidative stress. Biomed Res Int. 2014;2014:245210. doi: 10.1155/2014/245210 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Youngchaiyud U, Coates AS, Whittingham S, Mackay IR. Cellular-immune response to myelin protein: absence in multiple sclerosis and presence in cerebrovascular accidents. Aust N Z J Med. 1974;4:535–8. doi: 10.1111/j.1445-5994.1974.tb03233.x [DOI] [PubMed] [Google Scholar]
- 186.Singh V, Sadler R, Heindl S, Llovera G, Roth S, Benakis C, Liesz A. The gut microbiome primes a cerebroprotective immune response after stroke. J Cereb Blood Flow Metab. 2018;38:1293–1298. doi: 10.1177/0271678X18780130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Ma Q, Ineichen BV., Detmar M, Proulx ST. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat Commun. 2017;8:1434. doi: 10.1038/s41467-017-01484-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Javidi E, Magnus T. Autoimmunity After Ischemic Stroke and Brain Injury. Front Immunol. 2019;10:686. doi: 10.3389/fimmu.2019.00686 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Doyle KP, Quach LN, Solé M, Axtell RC, Nguyen TV V., Soler-Llavina GJ, Jurado S, Han J, Steinman L, Longo FM, et al. B-lymphocyte-mediated delayed cognitive impairment following stroke. J Neurosci. 2015;35:2133–2145. doi: 10.1523/JNEUROSCI.4098-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Xia G-H, You C, Gao X-X, Zeng X-L, Zhu J-J, Xu K-Y, Tan C-H, Xu R-T, Wu Q-H, Zhou H-W, et al. Stroke Dysbiosis Index (SDI) in Gut Microbiome Are Associated With Brain Injury and Prognosis of Stroke. Front Neurol. 2019;10:397. doi: 10.3389/fneur.2019.00397 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Yin J, Liao S-X, He Y, Wang S, Xia G-H, Liu F-T, Zhu J-J, You C, Chen Q, Zhou L, et al. Dysbiosis of Gut Microbiota With Reduced Trimethylamine-N-Oxide Level in Patients With Large-Artery Atherosclerotic Stroke or Transient Ischemic Attack. J Am Heart Assoc. 2015;4. doi: 10.1161/JAHA.115.002699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Haak BW, Westendorp WF, van Engelen TSR, Brands X, Brouwer MC, Vermeij J-D, Hugenholtz F, Verhoeven A, Derks RJ, Giera M, et al. Disruptions of Anaerobic Gut Bacteria Are Associated with Stroke and Post-stroke Infection: a Prospective Case-Control Study. Transl Stroke Res. 2021;12:581–592. doi: 10.1007/s12975-020-00863-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Yamashiro K, Tanaka R, Urabe T, Ueno Y, Yamashiro Y, Nomoto K, Takahashi T, Tsuji H, Asahara T, Hattori N. Gut dysbiosis is associated with metabolism and systemic inflammation in patients with ischemic stroke. PLoS One. 12:e0171521. doi: 10.1371/journal.pone.0171521 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Xu K, Gao X, Xia G, Chen M, Zeng N, Wang S, You C, Tian X, Di H, Tang W, et al. Rapid gut dysbiosis induced by stroke exacerbates brain infarction in turn. Gut. 2021;doi: 10.1136/gutjnl-2020-323263 [DOI] [PubMed] [Google Scholar]
- 195.Winek K, Engel O, Koduah P, Heimesaat MM, Fischer A, Bereswill S, Dames C, Kershaw O, Gruber AD, Curato C, et al. Depletion of Cultivatable Gut Microbiota by Broad-Spectrum Antibiotic Pretreatment Worsens Outcome After Murine Stroke. Stroke. 2016;47:1354–63. doi: 10.1161/STROKEAHA.115.011800 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Singh V, Roth S, Llovera G, Sadler R, Garzetti D, Stecher B, Dichgans M, Liesz A. Microbiota Dysbiosis Controls the Neuroinflammatory Response after Stroke. J Neurosci. 2016;36:7428–40. doi: 10.1523/JNEUROSCI.1114-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Crapser J, Ritzel R, Verma R, Venna VR, Liu F, Chauhan A, Koellhoffer E, Patel A, Ricker A, Maas K, et al. Ischemic stroke induces gut permeability and enhances bacterial translocation leading to sepsis in aged mice. Aging (Albany NY). 2016;8:1049–63. doi: 10.18632/aging.100952 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Li H, Xiao J, Li X, Chen H, Kang D, Shao Y, Shen B, Zhu Z, Yin X, Xie L, et al. Low Cerebral Exposure Cannot Hinder the Neuroprotective Effects of Panax Notoginsenosides. Drug Metab Dispos. 2018;46:53–65. doi: 10.1124/dmd.117.078436 [DOI] [PubMed] [Google Scholar]
- 199.Rahmati H, Momenabadi S, Vafaei AA, Bandegi AR, Mazaheri Z, Vakili A. Probiotic supplementation attenuates hippocampus injury and spatial learning and memory impairments in a cerebral hypoperfusion mouse model. Mol Biol Rep. 2019;46:4985–4995. doi: 10.1007/s11033-019-04949-7 [DOI] [PubMed] [Google Scholar]
- 200.Zhong D-Y, Li L, Ma R-M, Deng Y-H. The Effect of Probiotics in Stroke Treatment. Evid Based Complement Alternat Med. 2021;2021:4877311. doi: 10.1155/2021/4877311 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Bose S, Ramesh V, Locasale JW. Acetate Metabolism in Physiology, Cancer, and Beyond. Trends Cell Biol. 2019;29:695–703. doi: 10.1016/j.tcb.2019.05.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Durgan DJ, Lee J, McCullough LD, Bryan RM. Examining the Role of the Microbiota-Gut-Brain Axis in Stroke. Stroke. 2019;50:2270–2277. doi: 10.1161/STROKEAHA.119.025140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Corrêa-Oliveira R, Fachi JL, Vieira A, Sato FT, Vinolo MAR. Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunol. 2016;5:e73. doi: 10.1038/cti.2016.17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Margolis KG, Cryan JF, Mayer EA. The Microbiota-Gut-Brain Axis: From Motility to Mood. Gastroenterology. 2021;160:1486–1501. doi: 10.1053/j.gastro.2020.10.066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Le Poul E, Loison C, Struyf S, Springael J-Y, Lannoy V, Decobecq M-E, Brezillon S, Dupriez V, Vassart G, Van Damme J, et al. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278:25481–9. doi: 10.1074/jbc.M301403200 [DOI] [PubMed] [Google Scholar]
- 206.Onyszkiewicz M, Gawrys-Kopczynska M, Konopelski P, Aleksandrowicz M, Sawicka A, Koźniewska E, Samborowska E, Ufnal M. Butyric acid, a gut bacteria metabolite, lowers arterial blood pressure via colon-vagus nerve signaling and GPR41/43 receptors. Pflugers Arch. 2019;471:1441–1453. doi: 10.1007/s00424-019-02322-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology. 2013;145:396-406.e1–10. doi: 10.1053/j.gastro.2013.04.056 [DOI] [PubMed] [Google Scholar]
- 208.Liu L, Li L, Min J, Wang J, Wu H, Zeng Y, Chen S, Chu Z. Butyrate interferes with the differentiation and function of human monocyte-derived dendritic cells. Cell Immunol. 277:66–73. doi: 10.1016/j.cellimm.2012.05.011 [DOI] [PubMed] [Google Scholar]
- 209.Van Immerseel F, Ducatelle R, De Vos M, Boon N, Van De Wiele T, Verbeke K, Rutgeerts P, Sas B, Louis P, Flint HJ. Butyric acid-producing anaerobic bacteria as a novel probiotic treatment approach for inflammatory bowel disease. J Med Microbiol. 2010;59:141–143. doi: 10.1099/jmm.0.017541-0 [DOI] [PubMed] [Google Scholar]
- 210.Li J, Zhao F, Wang Y, Chen J, Tao J, Tian G, Wu S, Liu W, Cui Q, Geng B, et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome. 2017;5:14. doi: 10.1186/s40168-016-0222-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Erny D, Hrabě de Angelis AL, Jaitin D, Wieghofer P, Staszewski O, David E, Keren-Shaul H, Mahlakoiv T, Jakobshagen K, Buch T, et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci. 2015;18:965–77. doi: 10.1038/nn.4030 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Quinn M, McMillin M, Galindo C, Frampton G, Pae HY, DeMorrow S. Bile acids permeabilize the blood brain barrier after bile duct ligation in rats via Rac1-dependent mechanisms. Dig Liver Dis. 2014;46:527–34. doi: 10.1016/j.dld.2014.01.159 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Rüttimann EB, Arnold M, Hillebrand JJ, Geary N, Langhans W. Intrameal hepatic portal and intraperitoneal infusions of glucagon-like peptide-1 reduce spontaneous meal size in the rat via different mechanisms. Endocrinology. 2009;150:1174–81. doi: 10.1210/en.2008-1221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Castellanos-Jankiewicz A, Guzmán-Quevedo O, Fénelon VS, Zizzari P, Quarta C, Bellocchio L, Tailleux A, Charton J, Fernandois D, Henricsson M, et al. Hypothalamic bile acid-TGR5 signaling protects from obesity. Cell Metab. 2021;33:1483–1492.e10. doi: 10.1016/j.cmet.2021.04.009 [DOI] [PubMed] [Google Scholar]
- 215.Guo C, Xie S, Chi Z, Zhang J, Liu Y, Zhang L, Zheng M, Zhang X, Xia D, Ke Y, et al. Bile Acids Control Inflammation and Metabolic Disorder through Inhibition of NLRP3 Inflammasome. Immunity. 2016;45:802–816. doi: 10.1016/j.immuni.2016.09.008 [DOI] [PubMed] [Google Scholar]
- 216.Huang L, Xu G, Zhang R, Wang Y, Ji J, Long F, Sun Y. Increased admission serum total bile acids can be associated with decreased 3-month mortality in patients with acute ischemic stroke. Lipids Health Dis. 2022;21:15. doi: 10.1186/s12944-021-01620-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Charach G, Karniel E, Novikov I, Galin L, Vons S, Grosskopf I, Charach L. Reduced bile acid excretion is an independent risk factor for stroke and mortality: A prospective follow-up study. Atherosclerosis. 2020;293:79–85. doi: 10.1016/j.atherosclerosis.2019.12.010 [DOI] [PubMed] [Google Scholar]
- 218.Quintana FJ, Sherr DH. Aryl hydrocarbon receptor control of adaptive immunity. Pharmacol Rev. 2013;65:1148–61. doi: 10.1124/pr.113.007823 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Yano JM, Yu K, Donaldson GP, Shastri GG, Ann P, Ma L, Nagler CR, Ismagilov RF, Mazmanian SK, Hsiao EY. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161:264–76. doi: 10.1016/j.cell.2015.02.047 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Brouns R, Verkerk R, Aerts T, De Surgeloose D, Wauters A, Scharpé S, De Deyn PP. The role of tryptophan catabolism along the kynurenine pathway in acute ischemic stroke. Neurochem Res. 2010;35:1315–22. doi: 10.1007/s11064-010-0187-2 [DOI] [PubMed] [Google Scholar]
- 221.Ormstad H, Verkerk R, Aass HCD, Amthor K-F, Sandvik L. Inflammation-induced catabolism of tryptophan and tyrosine in acute ischemic stroke. J Mol Neurosci. 2013;51:893–902. doi: 10.1007/s12031-013-0097-2 [DOI] [PubMed] [Google Scholar]
- 222.Cuartero MI, Ballesteros I, de la Parra J, Harkin AL, Abautret-Daly A, Sherwin E, Fernández-Salguero P, Corbí AL, Lizasoain I, Moro MA. L-kynurenine/aryl hydrocarbon receptor pathway mediates brain damage after experimental stroke. Circulation. 2014;130:2040–51. doi: 10.1161/CIRCULATIONAHA.114.011394 [DOI] [PubMed] [Google Scholar]
- 223.Chen W-C, Chang L-H, Huang S-S, Huang Y-J, Chih C-L, Kuo H-C, Lee Y-H, Lee I-H. Aryl hydrocarbon receptor modulates stroke-induced astrogliosis and neurogenesis in the adult mouse brain. J Neuroinflammation. 2019;16:187. doi: 10.1186/s12974-019-1572-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Papandreou C, Moré M, Bellamine A. Trimethylamine N-Oxide in Relation to Cardiometabolic Health-Cause or Effect? Nutrients. 2020;12. doi: 10.3390/nu12051330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Wang Z, Klipfell E, Bennett BJ, Koeth R, Levison BS, Dugar B, Feldstein AE, Britt EB, Fu X, Chung Y-M, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472:57–63. doi: 10.1038/nature09922 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Zhu S, Jiang Y, Xu K, Cui M, Ye W, Zhao G, Jin L, Chen X. The progress of gut microbiome research related to brain disorders. J Neuroinflammation. 2020;17:25. doi: 10.1186/s12974-020-1705-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Koeth RA, Wang Z, Levison BS, Buffa JA, Org E, Sheehy BT, Britt EB, Fu X, Wu Y, Li L, et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat Med. 2013;19:576–85. doi: 10.1038/nm.3145 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Tang WHW, Wang Z, Levison BS, Koeth RA, Britt EB, Fu X, Wu Y, Hazen SL. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N Engl J Med. 2013;368:1575–84. doi: 10.1056/NEJMoa1109400 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Witkowski M, Weeks TL, Hazen SL. Gut Microbiota and Cardiovascular Disease. Circ Res. 2020;127:553–570. doi: 10.1161/CIRCRESAHA.120.316242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Wang Z, Tang WHW, Buffa JA, Fu X, Britt EB, Koeth RA, Levison BS, Fan Y, Wu Y, Hazen SL. Prognostic value of choline and betaine depends on intestinal microbiota-generated metabolite trimethylamine-N-oxide. Eur Heart J. 2014;35:904–10. doi: 10.1093/eurheartj/ehu002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Zhu W, Romano KA, Li L, Buffa JA, Sangwan N, Prakash P, Tittle AN, Li XS, Fu X, Androjna C, et al. Gut microbes impact stroke severity via the trimethylamine N-oxide pathway. Cell Host Microbe. 2021;29:1199–1208.e5. doi: 10.1016/j.chom.2021.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Haghikia A, Li XS, Liman TG, Bledau N, Schmidt D, Zimmermann F, Kränkel N, Widera C, Sonnenschein K, Haghikia A, et al. Gut Microbiota-Dependent Trimethylamine N-Oxide Predicts Risk of Cardiovascular Events in Patients With Stroke and Is Related to Proinflammatory Monocytes. Arterioscler Thromb Vasc Biol. 2018;38:2225–2235. doi: 10.1161/ATVBAHA.118.311023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Witkowski M, Weeks TL, Hazen SL. Gut Microbiota and Cardiovascular Disease. Circ Res. 2020;127:553–570. doi: 10.1161/CIRCRESAHA.120.316242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103. doi: 10.1126/science.aan4236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Bosetti F, Koenig JI, Ayata C, Back SA, Becker K, Broderick JP, Carmichael ST, Cho S, Cipolla MJ, Corbett D, et al. Translational Stroke Research: Vision and Opportunities. Stroke. 2017;48:2632–2637. doi: 10.1161/STROKEAHA.117.017112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Lourbopoulos A, Mourouzis I, Xinaris C, Zerva N, Filippakis K, Pavlopoulos A, Pantos C. Translational Block in Stroke: A Constructive and “Out-of-the-Box” Reappraisal. Front Neurosci. 2021;15:652403. doi: 10.3389/fnins.2021.652403 [DOI] [PMC free article] [PubMed] [Google Scholar]